1
|
Fu Y, Timp W, Sedlazeck FJ. Computational analysis of DNA methylation from long-read sequencing. Nat Rev Genet 2025:10.1038/s41576-025-00822-5. [PMID: 40155770 DOI: 10.1038/s41576-025-00822-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 04/01/2025]
Abstract
DNA methylation is a critical epigenetic mechanism in numerous biological processes, including gene regulation, development, ageing and the onset of various diseases such as cancer. Studies of methylation are increasingly using single-molecule long-read sequencing technologies to simultaneously measure epigenetic states such as DNA methylation with genomic variation. These long-read data sets have spurred the continuous development of advanced computational methods to gain insights into the roles of methylation in regulating chromatin structure and gene regulation. In this Review, we discuss the computational methods for calling methylation signals, contrasting methylation between samples, analysing cell-type diversity and gaining additional genomic insights, and then further discuss the challenges and future perspectives of tool development for DNA methylation research.
Collapse
Affiliation(s)
- Yilei Fu
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Computer Science, Rice University, Houston, TX, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
2
|
Alum EU, Ikpozu EN, Offor CE, Igwenyi IO, Obaroh IO, Ibiam UA, Ukaidi CUA. RNA-based diagnostic innovations: A new frontier in diabetes diagnosis and management. Diab Vasc Dis Res 2025; 22:14791641251334726. [PMID: 40230050 DOI: 10.1177/14791641251334726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
Background/Objective: Diabetes mellitus (DM) remains a major global health challenge due to its chronic nature and associated complications. Traditional diagnostic approaches, though effective, often lack the sensitivity required for early-stage detection. Recent advancements in molecular biology have identified RNA molecules, particularly non-coding RNAs such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), as promising biomarkers for diabetes. This review aims to explore the role of RNA-based biomarkers in the diagnosis, prognosis, and management of diabetes, highlighting their potential to revolutionize diabetes care.Method: A comprehensive literature review was conducted using electronic databases including PubMed, Scopus, and Web of Science. Articles published up to 2024 were screened and analyzed to extract relevant findings related to RNA-based diagnostics in diabetes. Emphasis was placed on studies demonstrating clinical utility, mechanistic insights, and translational potential of RNA molecules.Results: Numerous RNA species, particularly miRNAs such as miR-375, miR-29, and lncRNAs like H19 and MEG3, exhibit altered expression patterns in diabetic patients. These molecules are involved in key regulatory pathways of glucose metabolism, insulin resistance, and β-cell function. Circulating RNAs are detectable in various biofluids, enabling non-invasive diagnostic approaches. Emerging technologies, including RNA sequencing and liquid biopsy platforms, have enhanced the sensitivity and specificity of RNA detection, fostering the development of novel diagnostic tools and personalized therapeutic strategies.Conclusion: RNA-based biomarkers hold significant promise in advancing early detection, risk stratification, and therapeutic monitoring in diabetes care. Despite current challenges such as standardization and clinical validation, the integration of RNA diagnostics into routine clinical practice could transform diabetes management, paving the way for precision medicine approaches. Further research and multi-center trials are essential to validate these biomarkers and facilitate their regulatory approval and clinical implementation.
Collapse
Affiliation(s)
- Esther Ugo Alum
- Department of Research and Publications, Kampala International University, Uganda
- Department of Biochemistry, Ebonyi State University, Abakaliki, Nigeria
| | | | | | | | - Israel Olusegun Obaroh
- Department of Biological and Environmental Sciences, School of Natural and Applied Sciences, Kampala International University, Uganda
| | - Udu Ama Ibiam
- Department of Biochemistry, Ebonyi State University, Abakaliki, Nigeria
- Department of Biochemistry, College of Science, Evangel University Akaeze, Abakaliki, Nigeria
| | - Chris U A Ukaidi
- College of Economics and Management, Kampala International University, Uganda
| |
Collapse
|
3
|
Yang TC, Tsai JP, Hsu H, Chen YC, Liaw YC, Hsu SY, Yang HJ, Liaw YP. Delving Into the Interaction Between Exercise and Diabetes on Methylation of the FKBP5 Gene. J Diabetes Res 2025; 2025:1162708. [PMID: 40017583 PMCID: PMC11865466 DOI: 10.1155/jdr/1162708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Objective: FKBP5 is a critical gene involved in regulating the hypothalamic-pituitary-adrenal (HPA) axis and stress response. Aberrant DNA methylation at FKBP5 cytosine-phosphate-guanine (CpG) sites, such as cg22363520 and cg00862770, has been implicated in mental health disorders and metabolic diseases, including Type 2 diabetes. Exercise is a modulator of DNA methylation and metabolic health. This study investigates the interaction between exercise, diabetes, and FKBP5 methylation at cg22363520 and cg00862770 and explores their implications for mental health and disease development. Materials and Methods: FKBP5 methylation levels at cg22363520 and cg00862770 were analyzed in a cohort stratified by diabetes and exercise. Multiple linear regression models assessed the main effects and interactions of exercise and diabetes on FKBP5 methylation, with further stratified analyses for site-specific effects. Results: Exercise and diabetes showed significant and site-specific effects on FKBP5 methylation at cg22363520 and cg00862770. At cg22363520, exercise significantly reduced methylation levels in nondiabetic participants (β = -0.00195, p = 0.0157), while no significant effect was observed in diabetic individuals. Conversely, at cg00862770, exercise significantly decreased methylation levels in diabetic participants (β = -0.00611, p = 0.0081), with no significant effect in the nondiabetic group. Diabetes itself was associated with increased FKBP5 methylation at both sites, particularly in individuals without regular exercise. Additionally, significant interaction effects between exercise and diabetes were identified for both cg22363520 (p = 0.0336) and cg00862770 (p = 0.0021), highlighting the interplay between metabolic status and physical activity in regulating FKBP5 methylation. Conclusion: This study demonstrates that the effects of exercise on FKBP5 methylation are site-specific and influenced by diabetes status. Exercise reduces methylation at cg22363520 in nondiabetics and at cg00862770 in diabetics, indicating its role in modulating epigenetic regulation of stress and metabolic pathways. These findings underscore the interplay between exercise, diabetes, and FKBP5 methylation, with potential implications for improving mental health and metabolic outcomes.
Collapse
Affiliation(s)
- Teng-Chi Yang
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- Department of Medical Affairs, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County, Taiwan
| | - Jen Pi Tsai
- Division of Nephrology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County, Taiwan
- School of Medicine, Tzu Chi University, Buddhist Tzu Chi School Foundation, Hualien County, Taiwan
| | - Honda Hsu
- School of Medicine, Tzu Chi University, Buddhist Tzu Chi School Foundation, Hualien County, Taiwan
- Division of Plastic Surgery, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi County, Taiwan
| | - Yen-Chung Chen
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- Department of Neurology, Changhua Christian Hospital, Changhua City, Taiwan
| | - Yi-Chia Liaw
- Neurological Institute, Taipei Veterans General Hospital, Taipei City, Taiwan
| | - Shu Yi Hsu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
| | - Hao Jan Yang
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- Institute of Medicine, Chug Shan Medical University, Taichung City, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung City, Taiwan
| |
Collapse
|
4
|
Ding Q, Wojeck B, Zinchuk A. Understanding the impact of night-to-night sleep variations on glucose regulation in healthy young adults: Insights from Ng et al. (2024). Sleep 2025; 48:zsae253. [PMID: 39460669 PMCID: PMC11807883 DOI: 10.1093/sleep/zsae253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Indexed: 10/28/2024] Open
Affiliation(s)
- Qinglan Ding
- School of Nursing, College of Health and Human Sciences, Purdue University, West Lafayette, IN, USA
| | - Brian Wojeck
- Section of Endocrinology, Yale School of Medicine, New Haven, CT, USA
| | - Andrey Zinchuk
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
5
|
Li W, Luo P, Chen Q, Cheng L, Gan L, Zhang F, Zhong H, Zheng L, Qian B. Epigenetic modifications in bladder cancer: crosstalk between DNA methylation and miRNAs. Front Immunol 2025; 16:1518144. [PMID: 39981244 PMCID: PMC11841399 DOI: 10.3389/fimmu.2025.1518144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
Bladder cancer (BC) is a malignant tumor characterized by a high incidence of urinary system diseases. The complex pathogenesis of BC has long been a focal point in medical research. With the robust development of epigenetics, the crucial role of epigenetic modifications in the occurrence and progression of BC has been elucidated. These modifications not only affect gene expression but also impact critical biological behaviors of tumor cells, including proliferation, differentiation, apoptosis, invasion, and metastasis. Notably, DNA methylation, an important epigenetic regulatory mechanism, often manifests as global hypomethylation or hypermethylation of specific gene promoter regions in BC. Alterations in this methylation pattern can lead to increased genomic instability, which profoundly influences the expression of proto-oncogenes and tumor suppressor genes. MiRNAs, as noncoding small RNAs, participate in various biological processes of BC by regulating target genes. Consequently, this work aims to explore the interaction mechanisms between DNA methylation and miRNAs in the occurrence and development of BC. Research has demonstrated that DNA methylation not only directly influences the expression of miRNA genes but also indirectly affects the maturation and functionality of miRNAs by modulating the methylation status of miRNA promoter regions. Simultaneously, miRNAs can regulate DNA methylation levels by targeting key enzymes such as DNA methyltransferases (DNMTs), thereby establishing a complex feedback regulatory network. A deeper understanding of the crosstalk mechanisms between DNA methylation and miRNAs in BC will contribute to elucidating the complexity and dynamics of epigenetic modifications in this disease, and may provide new molecular targets and strategies for the early diagnosis, treatment, and prognostic evaluation of BC.
Collapse
Affiliation(s)
- Wei Li
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Qi Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Le Cheng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Lifeng Gan
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Fangtao Zhang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Haidong Zhong
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| | - Liying Zheng
- Department of Graduate, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Biao Qian
- Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Key Laboratory of Urology and Andrology of Ganzhou, Ganzhou, Jiangxi, China
| |
Collapse
|
6
|
Lai L, Juntilla DL, Del M, Del C Gomez-Alonso M, Grallert H, Thorand B, Farzeen A, Rathmann W, Winkelmann J, Prokisch H, Gieger C, Herder C, Peters A, Waldenberger M. Longitudinal association between DNA methylation and type 2 diabetes: findings from the KORA F4/FF4 study. Cardiovasc Diabetol 2025; 24:19. [PMID: 39827095 PMCID: PMC11748594 DOI: 10.1186/s12933-024-02558-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) has been linked to changes in DNA methylation levels, which can, in turn, alter transcriptional activity. However, most studies for epigenome-wide associations between T2D and DNA methylation comes from cross-sectional design. Few large-scale investigations have explored these associations longitudinally over multiple time-points. METHODS In this longitudinal study, we examined data from the Cooperative Health Research in the Region of Augsburg (KORA) F4 and FF4 studies, conducted approximately seven years apart. Leucocyte DNA methylation was assessed using the Illumina EPIC and 450K arrays. Linear mixed-effects models were employed to identify significant associations between methylation sites and diabetes status, as well as with fasting plasma glucose (FPG), hemoglobin A1c (HbA1c), homoeostasis model assessment of beta cell function (HOMA-B), and homoeostasis model assessment of insulin resistance (HOMA-IR). Interaction effects between diabetes status and follow-up time were also examined. Additionally, we explored CpG sites associated with persistent prediabetes or T2D, as well as the progression from normal glucose tolerance (NGT) to prediabetes or T2D. Finally, we assessed the associations between the identified CpG sites and their corresponding gene expression levels. RESULTS A total of 3,501 observations from 2,556 participants, with methylation measured at least once across two visits, were included in the analyses. We identified 64 sites associated with T2D including 15 novel sites as well as known associations like those with the thioredoxin-interacting protein (TXNIP) and ATP-binding cassette sub-family G member 1 (ABCG1) genes. Of these, eight CpG sites exhibited different rates of annual methylation change between the NGT and T2D groups, and seven CpG sites were linked to the progression from NGT to prediabetes or T2D, including those annotated to mannosidase alpha class 2a member 2 (MAN2A2) and carnitine palmitoyl transferase 1 A (CPT1A). Longitudinal analysis revealed significant associations between methylation and FPG at 128 sites, HbA1c at 41 sites, and HOMA-IR at 57 sites. Additionally, we identified 104 CpG-transcript pairs in whole blood, comprising 40 unique CpG sites and 96 unique gene transcripts. CONCLUSIONS Our study identified novel differentially methylated loci linked to T2D as well as to changes in diabetes status through a longitudinal approach. We report CpG sites with different rates of annual methylation change and demonstrate that DNA methylation associated with T2D is linked to following transcriptional differences. These findings provide new insights into the molecular mechanisms of diabetes development.
Collapse
Affiliation(s)
- Liye Lai
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Pettenkofer School of Public Health, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany.
| | - Dave Laurence Juntilla
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Pettenkofer School of Public Health, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | - Monica Del
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Monica Del C Gomez-Alonso
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Harald Grallert
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Pettenkofer School of Public Health, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Aiman Farzeen
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Juliane Winkelmann
- Institute of Human Genetics, School of Medicine, Technical University Munich, Munich, Germany
- Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
- Cluster for Systems Neurology (SyNergy), Munich, Germany
- Chair of Neurogenetics, Technische Universität München, Munich, Germany
| | - Holger Prokisch
- Institute of Human Genetics, School of Medicine, Technical University Munich, Munich, Germany
- Institute of Neurogenomics, Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Christian Herder
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute for Medical Information Processing, Biometry, and Epidemiology (IBE), Pettenkofer School of Public Health, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
7
|
Dall'Agnese A, Zheng MM, Moreno S, Platt JM, Hoang AT, Kannan D, Dall'Agnese G, Overholt KJ, Sagi I, Hannett NM, Erb H, Corradin O, Chakraborty AK, Lee TI, Young RA. Proteolethargy is a pathogenic mechanism in chronic disease. Cell 2025; 188:207-221.e30. [PMID: 39610243 PMCID: PMC11724756 DOI: 10.1016/j.cell.2024.10.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 08/07/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
The pathogenic mechanisms of many diseases are well understood at the molecular level, but there are prevalent syndromes associated with pathogenic signaling, such as diabetes and chronic inflammation, where our understanding is more limited. Here, we report that pathogenic signaling suppresses the mobility of a spectrum of proteins that play essential roles in cellular functions known to be dysregulated in these chronic diseases. The reduced protein mobility, which we call proteolethargy, was linked to cysteine residues in the affected proteins and signaling-related increases in excess reactive oxygen species. Diverse pathogenic stimuli, including hyperglycemia, dyslipidemia, and inflammation, produce similar reduced protein mobility phenotypes. We propose that proteolethargy is an overlooked cellular mechanism that may account for various pathogenic features of diverse chronic diseases.
Collapse
Affiliation(s)
| | - Ming M Zheng
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shannon Moreno
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jesse M Platt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - An T Hoang
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Deepti Kannan
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Kalon J Overholt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ido Sagi
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Nancy M Hannett
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Hailey Erb
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Olivia Corradin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arup K Chakraborty
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Tong Ihn Lee
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
8
|
Kumar S, Bhatia Z, Seshadri S. Formulated chitosan microspheres remodelled the altered gut microbiota and liver miRNA in diet-induced Type-2 diabetic rats. Carbohydr Res 2025; 547:109301. [PMID: 39580871 DOI: 10.1016/j.carres.2024.109301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024]
Abstract
Chitosan was formulated into a microsphere and comprehensively characterized and evaluated for its anti-inflammatory potential and anti-diabetic properties against the high sugar fat diet-induced diabetic animals. The diabetic model was induced through feeding with a high-sugar fat diet. Metformin, a standard antidiabetic drug, and CMS (chitosan microspheres) were administered orally for 90 days as reversal strategies. Upon completion of the study, the following parameters, such as serum biochemistry, cytokine analysis, tissue histology, liver miRNA sequencing, and Shotgun metagenomics studies from stool samples, were performed. SEM images of the microsphere indicated a smooth morphology, while FTIR and DSC respectively, confirmed the presence of functional groups of chitosan and the thermal stability of the formulation. Following HSFD induction, all the parameters analyzed were altered compared to the control group. In both reversal groups, serum biochemical parameters were restored, which was at par with the control. A significant increase in the anti-inflammatory cytokine IL-10, and a remarkable reduction in TNF-α and MCP-1 inflammatory cytokines were observed in both reversal groups. Tissue histology indicated improvements in low-grade inflammation, induced in the diabetic group. miR-203 was upregulated in the CMS-treated group, while miR-103 was downregulated. The study further delved into the impact on gut microbiota and KEGG. Major phyla i.e., Bacteroidetes, Cyanobacteria, Firmicutes, Proteobacteria, and Verrucomicrobia showed restoration, while upregulation of DNA polymerase zeta in T2D showed reversal after the treatment. The formulation showed reversal at par with metformin and also confirms its anti-diabetic and anti-inflammatory activities of CMS, with microfloral and miR regulatory functions.
Collapse
Affiliation(s)
- Sunny Kumar
- Institute of Science, Nirma University, Ahmedabad, India
| | - Zeel Bhatia
- Institute of Science, Nirma University, Ahmedabad, India
| | | |
Collapse
|
9
|
Guidi L, Martinez-Tellez B, Ortega Santos CP. Obesity, gut bacteria, and the epigenetic control of metabolic disease. NUTRITION IN THE CONTROL OF INFLAMMATION 2025:333-368. [DOI: 10.1016/b978-0-443-18979-1.00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Li Q. Correlation between lipid accumulation product and epigenetic age acceleration in American adults: a cross-sectional analysis using NHANES data. Eur J Med Res 2024; 29:575. [PMID: 39623450 PMCID: PMC11613462 DOI: 10.1186/s40001-024-02174-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND The risks of obesity and epigenetic age acceleration (EAA) have drawn widespread attention. Lipid accumulation product (LAP) is a simple and reliable indicator of obesity; however, the relationship between LAP and EAA remains unclear. METHODS Data from the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2002 were used. The EAA was assessed using a self-administered questionnaire in the database. LAP was calculated based on triglycerides and waist circumference. The association between LAP and EAA was analyzed using logistic regression models, subgroup analysis, and smooth curve fitting. RESULTS A total of 1796 participants were included in the study, of whom 1055 had EAA. After adjusting for relevant covariates, participants with EAA generally had higher LAP levels than those without EAA (258.1 vs. 244.6). Logistic regression analysis showed that individuals in the highest LAP quartile (Q4) were more likely to have EAA than those in the lowest quartile (Q1) (OR = 1.77; 95% CI 1.31-2.39; P < 0.001). The area under the curve of the adjusted logistic regression analysis was 0.706. CONCLUSION This research indicates that elevated LAP levels are independently linked to an increased risk of EAA, and early intervention to reduce high LAP levels is necessary to mitigate the progression of EAA.
Collapse
Affiliation(s)
- Qiqiang Li
- General Practice Department, Fuyong People's Hospital of Baoan District, Shenzhen, 518103, China.
| |
Collapse
|
11
|
Silva BCE, Drzewiecka EM, Piotrowska-Tomala K, Alpoim-Moreira J, Sadowska A, Kowalik MK, Pimenta J, Rebordão MR, Ferreira-Dias G, Skarzynski D, Szóstek-Mioduchowska A. The alteration in myometrial mRNA transcription of the regulatory genes of DNA methylation in mare with endometrosis. Reprod Biol 2024; 24:100962. [PMID: 39442277 DOI: 10.1016/j.repbio.2024.100962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/19/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
A reduction in myometrial contractile activity can lead to inadequate cleaning of the uterine lumen, resulting in persistent endometritis and potentially endometrosis in mares. Oxytocin (OXT) is a key hormonal regulator of myometrial contraction. While epigenetic regulation of myometrial gene expression has been studied in humans, there is limited information on the expression of DNA methyltransferases (DNMTs) and ten-eleven translocation enzymes (TETs) in the myometrium of mares. This study aimed to evaluate the mRNA transcription of these enzymes and the potential role of DNA methylation in the expression of the OXT receptor (OXTR) gene in the myometrium of mares with endometrosis. Myometrial samples were collected post-mortem during the mid-luteal (n = 23) and follicular (n = 20) phases of the estrous cycle and assessed according to Kenney and Doig endometrial category (I, IIA, IIB, III). mRNA transcription of OXTR, DNMT1, -3A, -3B and TET1, -2, -3 were determined using qPCR. DNA methylation analysis at CpG islands of OXTR exons 1 and 2 was performed using bisulfite pyrosequencing. Myometrial OXTR mRNA transcription and DNA methylation in its promoter region showed no significant differences between categories, although increased methylation was observed at CpG island position 6 in exon 2. DNMT1, TET2, and TET3 mRNA transcription was altered in the equine myometrium depending on the phase of the estrous cycle and the severity of endometrosis (P < 0.05). These findings indicate that DNMTs and TETs were expressed in myometrium in a manner specific to the severity of endometrosis and phases of the estrous cycle, suggesting a potential regulatory role in DNA methylation of myometrial gene expression.
Collapse
Affiliation(s)
| | - Ewa Monika Drzewiecka
- Institute of Animal Reproduction and Food Research Polish Academy of Sciences, 10-748 Olsztyn, Poland
| | | | - Joana Alpoim-Moreira
- C.I.I.S.A., Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Portugal
| | - Agnieszka Sadowska
- Institute of Animal Reproduction and Food Research Polish Academy of Sciences, 10-748 Olsztyn, Poland
| | | | - Jorge Pimenta
- C.I.I.S.A., Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Maria Rosa Rebordão
- C.I.I.S.A., Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Portugal; Polytechnic University of Coimbra, Coimbra Agriculture School, Bencanta, 3045-601 Coimbra, Portugal; Research Center for Natural Resources, Environment (CERNAS), Polytechnic University of Coimbra, Bencanta, 3045-601 Coimbra, Portugal
| | - Graça Ferreira-Dias
- C.I.I.S.A., Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Portugal
| | - Dariusz Skarzynski
- Institute of Animal Reproduction and Food Research Polish Academy of Sciences, 10-748 Olsztyn, Poland
| | - Anna Szóstek-Mioduchowska
- Institute of Animal Reproduction and Food Research Polish Academy of Sciences, 10-748 Olsztyn, Poland.
| |
Collapse
|
12
|
Tang Z, Zhao L, Li J, Yang Y, Liu F, Li H, Yang Z, Qin S, Li X. Prognostic effectiveness of interactive vs. non-interactive mobile app interventions in type 2 diabetes: a systematic review and meta-analysis. Arch Public Health 2024; 82:221. [PMID: 39578910 PMCID: PMC11583391 DOI: 10.1186/s13690-024-01450-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Mobile app interventions are emerging as significant tools in managing the prognosis of type 2 diabetes, demonstrating progressively greater impacts. The effectiveness of these interventions needs further evidence-based support. OBJECTIVE This study conducted a systematic review and meta-analysis of randomized controlled trials to evaluate the effectiveness of mobile app interventions in improving prognosis for patients with type 2 diabetes. METHODS We searched PubMed, Cochrane, Embase, and Web of Science for relevant studies published from inception to 18 April 2024, adhering to the Cochrane Handbook guidelines. The quality of the included studies was assessed using the Cochrane risk of bias tool. Primary outcomes measured were changes in glycated hemoglobin (HbA1c) and diabetes self-management (DSM). Secondary outcomes included changes in diastolic blood pressure (DBP), systolic blood pressure (SBP), triglycerides(TG), total cholesterol(TC), high-density lipoprotein (HDL), low-density lipoprotein (LDL), lipid profiles, fasting plasma glucose (FPG), body mass index (BMI), and Steps outcomes. Subgroup analyses were performed for the primary outcomes and for high-density lipoprotein (HDL), low-density lipoprotein (LDL), diastolic blood pressure (DBP), and systolic blood pressure (SBP). Interventions with or without interactions were also used as a basis for subgrouping. RESULTS A total of 15 eligible articles involving 17 studies with 2,028 subjects (1,123 in the intervention group and 1,020 in the control group) were included in the synthesis. Interactive mobile app interventions significantly reduced HbA1c levels (SMD - 0.24; 95% CI, -0.33 to -0.15; P < 0.00001) and significantly improved diabetes self-care (SMD 0.71; 95% CI, 0.21 to 1.21; P = 0.005). Secondary outcomes, including FPG, LDL, DBP, and SBP, showed varying degrees of improvement. Subgroup analyses indicated that the intervention effect was more pronounced and less heterogeneous in the short-term (≤ 3 months) for younger Asian individuals (< 55 years) who used an interactive mobile app. CONCLUSION Interactive mobile app interventions effectively improve HbA1c levels and diabetes self-care competencies in patients with type 2 diabetes. These interventions offer supportive evidence for their clinical use in managing and prognosticating type 2 diabetes. SYSTEMATIC REVIEW REGISTRATION CRD42024550643.
Collapse
Affiliation(s)
- Zheng Tang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Lijuan Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Jixin Li
- Xi yuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yang Yang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Fengzhao Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Han Li
- Department of Geriatric Medicine, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, Shandong, 250014, China
| | - Zhenyu Yang
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, China
| | - Shanyu Qin
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250014, China
| | - Xinqin Li
- Shandong Vocational College of Special Education, Jinan, Shandong, 250000, China.
- Department of acupuncture and moxibustion, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Shandong, 250014, Jinan, China.
| |
Collapse
|
13
|
Takenaka Y, Watanabe M. Environmental Factor Index (EFI): A Novel Approach to Measure the Strength of Environmental Influence on DNA Methylation in Identical Twins. EPIGENOMES 2024; 8:44. [PMID: 39584967 PMCID: PMC11587003 DOI: 10.3390/epigenomes8040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES The dynamic interaction between genomic DNA, epigenetic modifications, and phenotypic traits was examined in identical twins. Environmental perturbations can induce epigenetic changes in DNA methylation, influencing gene expression and phenotypes. Although DNA methylation mediates gene-environment correlations, the quantitative effects of external factors on DNA methylation remain underexplored. This study aimed to quantify these effects using a novel approach. METHODS A cohort study was conducted on healthy monozygotic twins to evaluate the influence of environmental stimuli on DNA methylation. We developed the Environmental Factor Index (EFI) to identify methylation sites showing statistically significant changes in response to environmental stimuli. We analyzed the identified sites for associations with disorders, DNA methylation markers, and CpG islands. RESULTS The EFI identified methylation sites that exhibited significant associations with genes linked to various disorders, particularly cancer. These sites were overrepresented on CpG islands compared to other genomic features, highlighting their regulatory importance. CONCLUSIONS The EFI is a valuable tool for understanding the molecular mechanisms underlying disease pathogenesis. It provides insights into the development of preventive and therapeutic strategies and offers a new perspective on the role of environmental factors in epigenetic regulation.
Collapse
Affiliation(s)
- Yoichi Takenaka
- Faculty of Informatics, Kansai University, Osaka 569-1052, Japan
- Center for Twin Research, Graduate School of Medicine, The University of Osaka, Osaka 565-0871, Japan (M.W.)
| | - Osaka Twin Research Group
- Center for Twin Research, Graduate School of Medicine, The University of Osaka, Osaka 565-0871, Japan (M.W.)
| | - Mikio Watanabe
- Center for Twin Research, Graduate School of Medicine, The University of Osaka, Osaka 565-0871, Japan (M.W.)
- Department of Clinical Laboratory and Biomedical Sciences, Graduate School of Medicine, The University of Osaka, Osaka 565-0871, Japan
| |
Collapse
|
14
|
Pool KD, Hemmat GJ, Dorschner RA. ECRG4 mediates host response to cutaneous infection by regulating neutrophil recruitment and adhesion receptor expression. PLoS One 2024; 19:e0310810. [PMID: 39509414 PMCID: PMC11542879 DOI: 10.1371/journal.pone.0310810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/04/2024] [Indexed: 11/15/2024] Open
Abstract
Rapid neutrophil recruitment is critical for controlling infection, with dysfunctional neutrophil responses in diseases like diabetes associated with greater morbidity and mortality. We have shown that the leukocyte protein ECRG4 enhances early neutrophil recruitment to cutaneous wounds and hypothesized that ECRG4 regulates the early host response to infection. Using a cutaneous infection model, we found that ECRG4 KO mice had decreased early neutrophil recruitment with persistent larger lesions, increased bacterial proliferation and systemic dissemination. Although previous work identified ECRG4 as a negative regulator of CD44 on neutrophils, the mechanism regulating neutrophil recruitment remained unknown. We demonstrated that pro-inflammatory responses were intact in ECRG4 KO mice, but found decreased neutrophil mobilization from bone marrow and decreased migration to chemokines. ECRG4 KO mouse neutrophils demonstrated an increase in adhesion molecules that regulate recruitment, including enhanced induction of integrin CD11b and increased L-selectin and CD44 on bone marrow neutrophils. Analysis of gene expression in leukocytes from diabetic patients found decreased ECRG4 expression with similar increased L-selectin and CD44. We propose a previously unrecognized mechanism governing neutrophil recruitment, whereby ECRG4 mediates neutrophil surface adhesion molecules that determine both recruitment and outside-in signaling that modulates neutrophil response to pro-inflammatory stimuli.
Collapse
Affiliation(s)
- Katie D. Pool
- Department of Dermatology, University of California San Diego, San Diego, CA, United States of America
- Division of Trauma, Surgical Critical Care and Burn, Department of Surgery, University of California San Diego, San Diego, CA, United States of America
| | - Gracie J. Hemmat
- Department of Dermatology, University of California San Diego, San Diego, CA, United States of America
| | - Robert A. Dorschner
- Department of Dermatology, University of California San Diego, San Diego, CA, United States of America
| |
Collapse
|
15
|
Roychaudhuri R, West T, Bhattacharya S, Saavedra HG, Lee H, Albacarys L, Gadalla MM, Amzel M, Yang P, Snyder SH. Mammalian D-Cysteine controls insulin secretion in the pancreas. Mol Metab 2024; 90:102043. [PMID: 39368613 PMCID: PMC11536007 DOI: 10.1016/j.molmet.2024.102043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND D-amino acids are being recognized as important molecules in mammals with function. This is a first identification of endogenous D-cysteine in mammalian pancreas. METHODS Using a novel stereospecific bioluminescent assay, chiral chromatography, enzyme kinetics and a transgenic mouse model we identify endogenous D-cysteine. We elucidate its function in two mice models of type 1 diabetes (STZ and NOD), and in tests of Glucose Stimulated Insulin Secretion in isolated mouse and human islets and INS-1 832/13 cell line. RESULTS AND DISCUSSION D-cysteine is synthesized by serine racemase (SR) and SR-/- mice produce 6-10 fold higher levels of insulin in the pancreas and plasma including higher glycogen and ketone bodies in the liver. The excess insulin is stored as amyloid in secretory vesicles and exosomes. In glucose stimulated insulin secretion in mouse and human islets, equimolar amount of D-cysteine showed higher inhibition of insulin secretion compared to D-serine, another closely related stereoisomer synthesized by SR. In mouse models of diabetes (Streptozotocin (STZ) and Non Obese Diabetes (NOD) and human pancreas, the diabetic state showed increased expression of D-cysteine compared to D-serine followed by increased expression of SR. SR-/- mice show decreased cAMP in the pancreas, lower DNA methyltransferase enzymatic and promoter activities followed by reduced phosphorylation of CREB (S133), resulting in decreased methylation of the Ins1 promoter. D-cysteine is efficiently metabolized by D-amino acid oxidase and transported by ASCT2 and Asc1. Dietary supplementation with methyl donors restored the high insulin levels and low DNMT enzymatic activity in SR-/- mice. CONCLUSIONS Our data show that endogenous D-cysteine in the mammalian pancreas is a regulator of insulin secretion.
Collapse
Affiliation(s)
- Robin Roychaudhuri
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Birth Defects, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Timothy West
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Soumyaroop Bhattacharya
- Department of Neonatology, University of Rochester Medical Center, Rochester, New York, NY 14642, USA
| | - Harry G Saavedra
- Centro de Investigacion en Bioingenieria, Universidad de Ingenieria y Tecnologia (UTEC), 15063 Lima, Peru
| | - Hangnoh Lee
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Lauren Albacarys
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Moataz M Gadalla
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mario Amzel
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Birth Defects, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Solomon H Snyder
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
16
|
Moghadasi M, Taherimoghaddam M, Babaeenezhad E, Birjandi M, Kaviani M, Moradi Sarabi M. MicroRNA-34a and promoter methylation contribute to peroxisome proliferator-activated receptor gamma gene expression in patients with type 2 diabetes. Diabetes Metab Syndr 2024; 18:103156. [PMID: 39522431 DOI: 10.1016/j.dsx.2024.103156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
AIMS This study aimed to investigate the roles of DNA methylation and miR-34a in the regulation of peroxisome proliferator-activated receptor gamma (PPARγ) in patients with type 2 diabetes (T2D). METHODS We investigated the methylation status of four regions of the PPARγ promoter and PPARγ expression in a panel of 84 T2D patients using methylation-specific PCR (MSP) and RT-qPCR, respectively. Moreover, we quantified DNA methyltransferases (DNMTs) expression and global DNA methylation levels by RT-qPCR and ELISA, respectively. We measured the expression levels of miR-34a and protein expression of PPARγ by stem-loop RT-qPCR and ELISA, respectively. RESULTS We found significant DNA hypermethylation in the R2 and R3 regions of the PPARγ promoter in people with diabetes. Functionally, this was associated with a significant reduction in PPARγ expression. In addition, we observed a significant increase in 5-methylcytosine levels in people with diabetes. A marked increase in circulating miR-34a in the early stages of T2D (up to 10 years) and a significant decrease in circulating miR-34a with increasing diabetes duration from 10 years after the onset of diabetes. Interestingly, upregulation of DNA methyltransferases 1 (DNMT1), DNMT3A, and DNMT3B was observed in people with diabetes, and the average expression of DNMTs was negatively correlated with circulating miR-34a levels. In contrast, the serum protein level of PPARγ, a direct target of miR-34a, increased considerably with diabetes duration and showed a negative correlation with circulating miR-34a, cholesterol, triglyceride, and low-density lipoprotein. CONCLUSION PPARγ promoter hypermethylation and miR-34a upregulation are associated with T2D pathogenesis through PPARγ dysregulation.
Collapse
Affiliation(s)
- Mona Moghadasi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mozhgan Taherimoghaddam
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Esmaeel Babaeenezhad
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mehdi Birjandi
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran; Department of Biostatistics and Epidemiology, School of Health and Nutrition, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mozhgan Kaviani
- Department of Internal Medicine, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mostafa Moradi Sarabi
- Department Clinical Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran; Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
17
|
Belin MAF, Vieira TA, Grandini NA, Siqueira JS, Palacio TLN, Cruzeiro J, Sormani LE, Tanganini MD, Barbosa GS, Gregolin CS, de Campos DHS, Bazan SGZ, Minatel IO, Lima GPP, Correa CR. Cardiac biogenic amine profile and its relationship with parameters of cardiovascular disease in obesity. Vascul Pharmacol 2024; 156:107412. [PMID: 39033868 DOI: 10.1016/j.vph.2024.107412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
AIMS To identify the cardiac biogenic amine profile of obese rats and associate these compounds with parameters of cardiovascular disease. MAIN METHODS Wistar rats (n = 20) were randomly distributed into two groups: control and obese. Obesity was induced by a high-sugar fat diet. Biochemical parameters were evaluated. Doppler Echocardiography and systolic blood pressure; interleukin-10 (IL-10), tumor necrosis factor-alpha (TNF-α), protein carbonylation, ferric reducing antioxidant power (FRAP), and catalase activity were measured in cardiac tissue. HPLC evaluated the cardiac biogenic profile. Data were compared using the Student's T or Mann-Whitney tests and Spearman's correlation at 5% significance. The principal component analysis (PCA) was performed. KEY FINDINGS Obesity generated hypertension, cardiac remodeling and dysfunction, and imbalanced all biochemical, inflammatory, and oxidative markers (p < 0.001). Eight biogenic amines were found in cardiac tissue. Obesity increased serotonin and decreased agmatine, putrescine, cadaverine, and spermidine. Serotonin (r = 0.534 to 0.808) was strong and positively correlated with obesity, biochemical parameters, cardiac inflammation, oxidative stress, hypertension, cardiac remodeling, and dysfunction (p < 0.001). Spermidine (r = -0.560 to -0.680), putrescine (r = -0.532 to -0.805), cadaverine (r = -0.534 to -0.860), and agmatine (r = -0.579 to -0.884) were inversely correlated with the same parameters (p < 0.001). PCA allowed for distinguishing the control and obese groups. SIGNIFICANCE There are strong correlations between cardiac biogenic amine levels, cardiac remodeling, and dysfunction resulting from obesity. CONCLUSION There is an association between cardiac biogenic amines and cardiovascular disease in obesity. In addition, agmatine, putrescine, cadaverine, and, mainly, serotonin may be new biomarkers for cardiovascular health in obesity and help to improve the diagnosis and treatment of CVD resulting or not from obesity. However, more research is needed to support this conclusion.
Collapse
Affiliation(s)
| | - Taynara Aparecida Vieira
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Núbia Alves Grandini
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Juliana Silva Siqueira
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | | | - Jordanna Cruzeiro
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Luis Eduardo Sormani
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Murilo Dalarme Tanganini
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Gabriela Souza Barbosa
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Cristina Schmitt Gregolin
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | | | | | - Igor Otávio Minatel
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Giuseppina Pace Pereira Lima
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University (Unesp), Botucatu 18618-687, Brazil
| | - Camila Renata Correa
- Department of Pathology, Medical School, Sao Paulo State University (Unesp), Botucatu 18618-687, Brazil.
| |
Collapse
|
18
|
Ray D, Loomis SJ, Venkataraghavan S, Zhang J, Tin A, Yu B, Chatterjee N, Selvin E, Duggal P. Characterizing Common and Rare Variations in Nontraditional Glycemic Biomarkers Using Multivariate Approaches on Multiancestry ARIC Study. Diabetes 2024; 73:1537-1550. [PMID: 38869630 PMCID: PMC11333373 DOI: 10.2337/db23-0318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Genetic studies of nontraditional glycemic biomarkers, glycated albumin and fructosamine, can shed light on unknown aspects of type 2 diabetes genetics and biology. We performed a multiphenotype genome-wide association study of glycated albumin and fructosamine from 7,395 White and 2,016 Black participants in the Atherosclerosis Risk in Communities (ARIC) study on common variants from genotyped/imputed data. We discovered two genome-wide significant loci, one mapping to a known type 2 diabetes gene (ARAP1/STARD10) and another mapping to a novel region (UGT1A complex of genes), using multiomics gene-mapping strategies in diabetes-relevant tissues. We identified additional loci that were ancestry- and sex-specific (e.g., PRKCA in African ancestry, FCGRT in European ancestry, TEX29 in males). Further, we implemented multiphenotype gene-burden tests on whole-exome sequence data from 6,590 White and 2,309 Black ARIC participants. Ten variant sets annotated to genes across different variant aggregation strategies were exome-wide significant only in multiancestry analysis, of which CD1D, EGFL7/AGPAT2, and MIR126 had notable enrichment of rare predicted loss of function variants in African ancestry despite smaller sample sizes. Overall, 8 of 14 discovered loci and genes were implicated to influence these biomarkers via glycemic pathways, and most of them were not previously implicated in studies of type 2 diabetes. This study illustrates improved locus discovery and potential effector gene discovery by leveraging joint patterns of related biomarkers across the entire allele frequency spectrum in multiancestry analysis. Future investigation of the loci and genes potentially acting through glycemic pathways may help us better understand the risk of developing type 2 diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Debashree Ray
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | | | - Sowmya Venkataraghavan
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Jiachen Zhang
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Adrienne Tin
- School of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX
| | - Nilanjan Chatterjee
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Elizabeth Selvin
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Welch Center for Prevention, Epidemiology, & Clinical Research, Johns Hopkins University, Baltimore, MD
| | - Priya Duggal
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
19
|
Svikle Z, Paramonova N, Siliņš E, Pahirko L, Zariņa L, Baumane K, Petrovski G, Sokolovska J. DNA Methylation Profiles of PSMA6, PSMB5, KEAP1, and HIF1A Genes in Patients with Type 1 Diabetes and Diabetic Retinopathy. Biomedicines 2024; 12:1354. [PMID: 38927561 PMCID: PMC11202151 DOI: 10.3390/biomedicines12061354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
We explored differences in the DNA methylation statuses of PSMA6, PSMB5, HIF1A, and KEAP1 gene promoter regions in patients with type 1 diabetes and different diabetic retinopathy (DR) stages. Study subjects included individuals with no DR (NDR, n = 41), those with non-proliferative DR (NPDR, n = 27), and individuals with proliferative DR or those who underwent laser photocoagulation (PDR/LPC, n = 46). DNA methylation was determined by Zymo OneStep qMethyl technique. The methylation of PSMA6 (NDR 5.9 (3.9-8.7) %, NPDR 4.5 (3.8-5.7) %, PDR/LPC 6.6 (4.7-10.7) %, p = 0.003) and PSMB5 (NDR 2.2 (1.9-3.7) %, NPDR 2.2 (1.9-3.0) %, PDR/LPC 3.2 (2.5-7.1) %, p < 0.01) differed across the groups. Consistent correlations were observed between the methylation levels of HIF1A and PSMA6 in all study groups. DNA methylation levels of PSMA6, PSMB5, and HIF1A genes were positively correlated with the duration of diabetes, HbA1c, and albuminuria in certain study groups. Univariate regression models revealed a significant association between the methylation level z-scores of PSMA6, PSMB5, and HIF1A and severe DR (PSMA6: OR = 1.96 (1.15; 3.33), p = 0.013; PSMB5: OR = 1.90 (1.14; 3.16), p = 0.013; HIF1A: OR = 3.19 (1.26; 8.06), p = 0.014). PSMB5 remained significantly associated with DR in multivariate analysis. Our findings suggest significant associations between the severity of DR and the DNA methylation levels of the genes PSMA6, PSMB5, and HIF1A, but not KEAP1 gene.
Collapse
Affiliation(s)
- Zane Svikle
- Faculty of Medicine, University of Latvia, Jelgavas Street 3, LV 1004 Riga, Latvia; (Z.S.); (L.Z.); (K.B.)
| | - Natalia Paramonova
- Institute of Biology, University of Latvia, Jelgavas Street 1, LV 1004 Riga, Latvia;
| | - Emīls Siliņš
- Faculty of Physics, Mathematics and Optometry, University of Latvia, Jelgavas Street 3, LV 1004 Riga, Latvia; (E.S.); (L.P.)
| | - Leonora Pahirko
- Faculty of Physics, Mathematics and Optometry, University of Latvia, Jelgavas Street 3, LV 1004 Riga, Latvia; (E.S.); (L.P.)
| | - Līga Zariņa
- Faculty of Medicine, University of Latvia, Jelgavas Street 3, LV 1004 Riga, Latvia; (Z.S.); (L.Z.); (K.B.)
- Ophthalmology Department, Riga East University Hospital, Hipokrata Street 2, LV 1038 Riga, Latvia
| | - Kristīne Baumane
- Faculty of Medicine, University of Latvia, Jelgavas Street 3, LV 1004 Riga, Latvia; (Z.S.); (L.Z.); (K.B.)
- Ophthalmology Department, Riga East University Hospital, Hipokrata Street 2, LV 1038 Riga, Latvia
| | - Goran Petrovski
- Center of Eye Research and Innovative Diagnostics, Department of Ophthalmology, Oslo University Hospital, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
| | - Jelizaveta Sokolovska
- Faculty of Medicine, University of Latvia, Jelgavas Street 3, LV 1004 Riga, Latvia; (Z.S.); (L.Z.); (K.B.)
| |
Collapse
|
20
|
Sánchez-Ortí JV, Correa-Ghisays P, Balanzá-Martínez V, Macías Saint-Gerons D, Berenguer-Pascual E, Romá-Mateo C, Victor VM, Forés-Martos J, San-Martin C, Selva-Vera G, Tabarés-Seisdedos R. Systemic inflammation, oxidative damage and neurocognition predict telomere length in a transdiagnostic sample stratified by global DNA methylation levels. Sci Rep 2024; 14:13159. [PMID: 38849401 PMCID: PMC11161596 DOI: 10.1038/s41598-024-62980-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Epigenetic mechanisms contribute to the maintenance of both type 2 diabetes mellitus (T2DM) and psychiatric disorders. Emerging evidence suggests that molecular pathways and neurocognitive performance regulate epigenetic dynamics in these disorders. The current combined and transdiagnostic study investigated whether inflammatory, oxidative stress, adhesion molecule, neurocognitive and functional performance are significant predictors of telomere dynamics in a sample stratified by global DNA methylation levels. Peripheral blood inflammation, oxidative stress and adhesion molecule biomarkers and neurocognitive function were assessed twice over a 1-year period in 80 individuals, including 16 with schizophrenia (SZ), 16 with bipolar disorder (BD), 16 with major depressive disorder (MDD), 15 with T2DM, and 17 healthy controls (HCs). Leukocyte telomere length (LTL) was measured by qRT-PCR using deoxyribonucleic acid (DNA) extracted from peripheral blood samples. A posteriori, individuals were classified based on their global methylation score (GMS) at baseline into two groups: the below-average methylation (BM) and above-average methylation (AM) groups. Hierarchical and k-means clustering methods, mixed one-way analysis of variance and linear regression analyses were performed. Overall, the BM group showed a significantly higher leukocyte telomere length (LTL) than the AM group at both time points (p = 0.02; η2p = 0.06). Moreover, the BM group had significantly lower levels of tumor necrosis factor alpha (TNF-α) (p = 0.03; η2p = 0.06) and C-reactive protein (CRP) (p = 0.03; η2p = 0.06) than the AM group at the 1-year follow-up. Across all participants, the regression models showed that oxidative stress (reactive oxygen species [ROS]) (p = 0.04) and global cognitive score [GCS] (p = 0.02) were significantly negatively associated with LTL, whereas inflammatory (TNF-α) (p = 0.04), adhesion molecule biomarkers (inter cellular adhesion molecule [ICAM]) (p = 0.009), and intelligence quotient [IQ] (p = 0.03) were significantly positively associated with LTL. Moreover, the model predictive power was increased when tested in both groups separately, explaining 15.8% and 28.1% of the LTL variance at the 1-year follow-up for the AM and BM groups, respectively. Heterogeneous DNA methylation in individuals with T2DM and severe mental disorders seems to support the hypothesis that epigenetic dysregulation occurs in a transdiagnostic manner. Our results may help to elucidate the interplay between epigenetics, molecular processes and neurocognitive function in these disorders. DNA methylation and LTL are potential therapeutic targets for transdiagnostic interventions to decrease the risk of comorbidities.
Collapse
Affiliation(s)
- Joan Vicent Sánchez-Ortí
- INCLIVA - Health Research Institute, Valencia, Spain
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain
- Faculty of Psychology, University of Valencia, Valencia, Spain
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain
| | - Patricia Correa-Ghisays
- INCLIVA - Health Research Institute, Valencia, Spain
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain
- Faculty of Psychology, University of Valencia, Valencia, Spain
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain
| | - Vicent Balanzá-Martínez
- INCLIVA - Health Research Institute, Valencia, Spain.
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain.
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain.
- Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain.
- VALSME (VALencia Salut Mental i Estigma), University of Valencia, Valencia, Spain.
| | - Diego Macías Saint-Gerons
- INCLIVA - Health Research Institute, Valencia, Spain
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain
| | | | - Carlos Romá-Mateo
- INCLIVA - Health Research Institute, Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), Health Institute, Carlos III, Madrid, Spain
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Víctor M Victor
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
- Service of Endocrinology and Nutrition, University Hospital Dr. Peset, Valencia, Spain
- Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain
| | - Jaume Forés-Martos
- INCLIVA - Health Research Institute, Valencia, Spain
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain
| | - Constanza San-Martin
- INCLIVA - Health Research Institute, Valencia, Spain
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain
- Department of Physiotherapy, University of Valencia, Valencia, Spain
| | - Gabriel Selva-Vera
- INCLIVA - Health Research Institute, Valencia, Spain
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain
- Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain
| | - Rafael Tabarés-Seisdedos
- INCLIVA - Health Research Institute, Valencia, Spain.
- TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain.
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain.
- Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain.
| |
Collapse
|
21
|
Wang X, Huangfu W, Zhao F. Correlation of ChREBP Gene Methylation with Pathological Characteristics of Type 2 Diabetes Mellitus. Appl Biochem Biotechnol 2024; 196:3076-3087. [PMID: 37615853 DOI: 10.1007/s12010-023-04714-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 08/25/2023]
Abstract
The objective of this study is to investigate the expression of the carbohydrate response element binding protein (ChREBP) gene in type 2 diabetes mellitus (T2DM) and its correlation with pathological features. For obtaining and exploring the pathological features in patients, sixty T2DM patients (the research group) and thirty healthy controls (the control group) presented to our hospital between January 2019 and June 2019 were selected as the research participants. After DNA extraction from peripheral blood mononuclear cells (PBMCs) and modification of target gene methylation with bisulfite, differences in methylation were verified, and the correlation of ChREBP methylation level with T2DM pathological features and single nucleotide polymorphism (SNP) typing was discussed. According to the prediction results of UCSC Genome Browser Home, there were two CpG islands in the promoter region of the ChREBP gene, and the first exon was selected for research. The ChREBP methylation rate was statistically higher in the research group versus the control group (P < 0.05). Age, FPG, TC, and TG were confirmed by the multiple linear regression analysis to be correlated with the ChREBP methylation rate (P < 0.05). Finally, there was no difference in ChREBP methylation level between CT- and CC-type patients at rs17145750 and rs1051921 loci (P > 0.05). Peripheral blood ChREBP methylation is elevated in T2DM patients and is closely related to age, blood glucose, and blood-lipid level, which is expected to be a new direction for future T2DM diagnosis and treatment.
Collapse
Affiliation(s)
- Xin Wang
- Department of General Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010000, China.
| | - Weizhong Huangfu
- Department of General Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010000, China
| | - Feng Zhao
- Department of General Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010000, China
| |
Collapse
|
22
|
Singh S, Kriti M, K.S. A, Sarma DK, Verma V, Nagpal R, Mohania D, Tiwari R, Kumar M. Deciphering the complex interplay of risk factors in type 2 diabetes mellitus: A comprehensive review. Metabol Open 2024; 22:100287. [PMID: 38818227 PMCID: PMC11137529 DOI: 10.1016/j.metop.2024.100287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024] Open
Abstract
The complex and multidimensional landscape of type 2 diabetes mellitus (T2D) is a major global concern. Despite several years of extensive research, the precise underlying causes of T2D remain elusive, but evidence suggests that it is influenced by a myriad of interconnected risk factors such as epigenetics, genetics, gut microbiome, environmental factors, organelle stress, and dietary habits. The number of factors influencing the pathogenesis is increasing day by day which worsens the scenario; meanwhile, the interconnections shoot up the frame. By gaining deeper insights into the contributing factors, we may pave the way for the development of personalized medicine, which could unlock more precise and impactful treatment pathways for individuals with T2D. This review summarizes the state of knowledge about T2D pathogenesis, focusing on the interplay between various risk factors and their implications for future therapeutic strategies. Understanding these factors could lead to tailored treatments targeting specific risk factors and inform prevention efforts on a population level, ultimately improving outcomes for individuals with T2D and reducing its burden globally.
Collapse
Affiliation(s)
- Samradhi Singh
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| | - Mona Kriti
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| | - Anamika K.S.
- Christ Deemed to Be University Bangalore, Karnataka, India
| | - Devojit Kumar Sarma
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, 226014, Uttar Pradesh, India
| | - Ravinder Nagpal
- Department of Nutrition & Integrative Physiology, College of Health & Human Sciences, Florida State University, Tallahassee, FL, 32306, USA
| | - Dheeraj Mohania
- Dr. R. P. Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Rajnarayan Tiwari
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| | - Manoj Kumar
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhauri, Bhopal, 462030, Madhya Pradesh, India
| |
Collapse
|
23
|
Salama OE, Hizon N, Del Vecchio M, Kolsun K, Fonseca MA, Lin DTS, Urtatiz O, MacIsaac JL, Kobor MS, Sellers EAC, Dolinsky VW, Dart AB, Jones MJ, Wicklow BA. DNA methylation signatures of youth-onset type 2 diabetes and exposure to maternal diabetes. Clin Epigenetics 2024; 16:65. [PMID: 38741114 DOI: 10.1186/s13148-024-01675-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
OBJECTIVE Youth-onset type 2 diabetes (T2D) is physiologically distinct from adult-onset, but it is not clear how the two diseases differ at a molecular level. In utero exposure to maternal type 2 diabetes (T2D) is known to be a specific risk factor for youth-onset T2D. DNA methylation (DNAm) changes associated with T2D but which differ between youth- and adult-onset might delineate the impacts of T2D development at different ages and could also determine the contribution of exposure to in utero diabetes. METHODS We performed an epigenome-wide analysis of DNAm on whole blood from 218 youth with T2D and 77 normoglycemic controls from the iCARE (improving renal Complications in Adolescents with type 2 diabetes through REsearch) cohort. Associations were tested using multiple linear regression models while adjusting for maternal diabetes, sex, age, BMI, smoking status, second-hand smoking exposure, cell-type proportions and genetic ancestry. RESULTS We identified 3830 differentially methylated sites associated with youth T2D onset, of which 3794 were moderately (adjusted p-value < 0.05 and effect size estimate > 0.01) associated and 36 were strongly (adjusted p-value < 0.05 and effect size estimate > 0.05) associated. A total of 3725 of these sites were not previously reported in the EWAS Atlas as associated with T2D, adult obesity or youth obesity. Moreover, three CpGs associated with youth-onset T2D in the PFKFB3 gene were also associated with maternal T2D exposure (FDR < 0.05 and effect size > 0.01). This is the first study to link PFKFB3 and T2D in youth. CONCLUSION Our findings support that T2D in youth has different impacts on DNAm than adult-onset, and suggests that changes in DNAm could provide an important link between in utero exposure to maternal diabetes and the onset of T2D.
Collapse
Affiliation(s)
- Ola E Salama
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Nikho Hizon
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Melissa Del Vecchio
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Kurt Kolsun
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Mario A Fonseca
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - David T S Lin
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
| | - Oscar Urtatiz
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
| | - Julia L MacIsaac
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
| | - Michael S Kobor
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Centre for Molecular Medicine and Therapeutics, Vancouver, BC, Canada
- Edwin S.H. Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth A C Sellers
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Vernon W Dolinsky
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - Allison B Dart
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | - Meaghan J Jones
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB, Canada.
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
| | - Brandy A Wicklow
- Diabetes Research Envision and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
24
|
Burghardt KJ, Burghardt PR, Howlett BH, Dass SE, Zahn B, Imam AA, Mallisho A, Msallaty Z, Seyoum B, Yi Z. Alterations in Skeletal Muscle Insulin Signaling DNA Methylation: A Pilot Randomized Controlled Trial of Olanzapine in Healthy Volunteers. Biomedicines 2024; 12:1057. [PMID: 38791018 PMCID: PMC11117943 DOI: 10.3390/biomedicines12051057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Antipsychotics are associated with severe metabolic side effects including insulin resistance; however, the mechanisms underlying this side effect are not fully understood. The skeletal muscle plays a critical role in insulin-stimulated glucose uptake, and changes in skeletal muscle DNA methylation by antipsychotics may play a role in the development of insulin resistance. A double-blind, placebo-controlled trial of olanzapine was performed in healthy volunteers. Twelve healthy volunteers were randomized to receive 10 mg/day of olanzapine for 7 days. Participants underwent skeletal muscle biopsies to analyze DNA methylation changes using a candidate gene approach for the insulin signaling pathway. Ninety-seven methylation sites were statistically significant (false discovery rate < 0.05 and beta difference between the groups of ≥10%). Fifty-five sites had increased methylation in the skeletal muscle of olanzapine-treated participants while 42 were decreased. The largest methylation change occurred at a site in the Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-Alpha (PPARGC1A) gene, which had 52% lower methylation in the olanzapine group. Antipsychotic treatment in healthy volunteers causes significant changes in skeletal muscle DNA methylation in the insulin signaling pathway. Future work will need to expand on these findings with expression analyses.
Collapse
Affiliation(s)
- Kyle J. Burghardt
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (B.H.H.); (S.E.D.)
| | - Paul R. Burghardt
- Department of Nutrition and Food Science, Wayne State University, Detroit, MI 48202, USA;
| | - Bradley H. Howlett
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (B.H.H.); (S.E.D.)
| | - Sabrina E. Dass
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (B.H.H.); (S.E.D.)
| | - Brent Zahn
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Ahmad A. Imam
- Internal Medicine Department, College of Medicine, Umm Al-Qura University, Makkah 24381, Saudi Arabia;
| | - Abdullah Mallisho
- Division of Endocrinology, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (A.M.); (Z.M.); (B.S.)
| | - Zaher Msallaty
- Division of Endocrinology, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (A.M.); (Z.M.); (B.S.)
| | - Berhane Seyoum
- Division of Endocrinology, School of Medicine, Wayne State University, Detroit, MI 48202, USA; (A.M.); (Z.M.); (B.S.)
| | - Zhengping Yi
- Department of Pharmaceutical Science, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48202, USA;
| |
Collapse
|
25
|
Ferenc K, Marcinkowski M, Olszewski J, Kowalczyk P, Pilžys T, Garbicz D, Dib N, Świderska B, Matyba P, Gajewski Z, Grzesiuk E, Zabielski R. The proteomic profile is altered but not repaired after bariatric surgery in type 2 diabetes pigs. Sci Rep 2024; 14:10235. [PMID: 38702370 PMCID: PMC11068747 DOI: 10.1038/s41598-024-60022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
To reveal the sources of obesity and type 2 diabetes (T2D) in humans, animal models, mainly rodents, have been used. Here, we propose a pig model of T2D. Weaned piglets were fed high fat/high sugar diet suppling 150% of metabolizable energy. Measurements of weight gain, blood morphology, glucose plasma levels, cholesterol, and triglycerides, as well as glucose tolerance (oral glucose tolerance test, OGTT) were employed to observe T2D development. The histology and mass spectrometry analyses were made post mortem. Within 6 months, the high fat-high sugar (HFHS) fed pigs showed gradual and significant increase in plasma triglycerides and glucose levels in comparison to the controls. Using OGTT test, we found stable glucose intolerance in 10 out of 14 HFHS pigs. Mass spectrometry analysis indicated significant changes in 330 proteins in the intestine, liver, and pancreas of the HFHS pigs. These pigs showed also an increase in DNA base modifications and elevated level of the ALKBH proteins in the tissues. Six diabetic HFHS pigs underwent Scopinaro bariatric surgery restoring glycaemia one month after surgery. In conclusion, a high energy diet applied to piglets resulted in the development of hyperlipidaemia, hyperglycaemia, and type 2 diabetes being reversed by a bariatric procedure, excluding the proteomic profile utill one month after the surgery.
Collapse
Affiliation(s)
- Karolina Ferenc
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland
| | - Michał Marcinkowski
- Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Jarosław Olszewski
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland
| | - Paweł Kowalczyk
- Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110, Jabłonna, Poland
| | - Tomaš Pilžys
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Damian Garbicz
- Institute of Oncology, Maria Sklodowska-Curie National Research, W.K. Roentgena 5, 02-781, Warsaw, Poland
| | - Naser Dib
- European Health Centre Otwock (ECZ Otwock), The Fryderyk Chopin Hospital, Borowa 14/18, 05-400, Otwock, Poland
| | - Bianka Świderska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Piotr Matyba
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland
| | - Zdzisław Gajewski
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland
| | - Elżbieta Grzesiuk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland.
| | - Romuald Zabielski
- Center for Translational Medicine, Warsaw University of Life Sciences, Nowoursynowska 100, 02-797, Warsaw, Poland.
| |
Collapse
|
26
|
Yang M, Wang M, Zhao X, Xu F, Liang S, Wang Y, Wang N, Sambou ML, Jiang Y, Dai J. DNA methylation marker identification and poly-methylation risk score in prediction of healthspan termination. Epigenomics 2024; 16:461-472. [PMID: 38482663 DOI: 10.2217/epi-2023-0343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024] Open
Abstract
Aim: To elucidate the epigenetic consequences of DNA methylation in healthspan termination (HST), considering the current limited understanding. Materials & methods: Genetically predicted DNA methylation models were established (n = 2478). These models were applied to genome-wide association study data on HST. Then, a poly-methylation risk score (PMRS) was established in 241,008 individuals from the UK Biobank. Results: Of the 63,046 CpGs from the prediction models, 13 novel CpGs were associated with HST. Furthermore, people with high PMRSs showed higher HST risk (hazard ratio: 1.18; 95% CI: 1.13-1.25). Conclusion: The study indicates that DNA methylation may influence HST by regulating the expression of genes (e.g., PRMT6, CTSK). PMRSs have a promising application in discriminating subpopulations to facilitate early prevention.
Collapse
Affiliation(s)
- Meiqi Yang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Mei Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaoyu Zhao
- Department of Statistics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Feifei Xu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Shuang Liang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yifan Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Nanxi Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Muhammed Lamin Sambou
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
| | - Yue Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention & Treatment, Collaborative Innovation Center for Cancer Personalized Medicine & China International Cooperation Center for Environment & Human Health, Gusu School, Nanjing Medical University, Nanjing, 211166, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention & Treatment, Collaborative Innovation Center for Cancer Personalized Medicine & China International Cooperation Center for Environment & Human Health, Gusu School, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
27
|
Muhammad A, Hixon JC, Pharmacy Yusuf A, Rivas Zarete JI, Johnson I, Miller J, Adu-Addai B, Yates C, Mahavadi S. Sex-specific epigenetics drive low GPER expression in gastrointestinal smooth muscles in type 2 diabetic mice. Sci Rep 2024; 14:5633. [PMID: 38453938 PMCID: PMC10920797 DOI: 10.1038/s41598-024-54213-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
Type 2 diabetes mellitus (T2D) causes gastroparesis, delayed intestinal transit, and constipation, for unknown reasons. Complications are predominant in women than men (particularly pregnant and postmenopausal women), suggesting a female hormone-mediated mechanism. Low G-protein coupled estrogen receptor (GPER) expression from epigenetic modifications may explain it. We explored sexually differentiated GPER expression and gastrointestinal symptoms related to GPER alterations in wild-type (WT) and T2D mice (db/db). We also created smooth muscle-specific GPER knockout (GPER KO) mice to phenotypically explore the effect of GPER deficiency on gastrointestinal motility. GPER mRNA and protein expression, DNA methylation and histone modifications were measured from stomach and colon samples of db/db and WT mice. Changes in gut motility were also evaluated as daily fecal pellet production patterns. We found that WT female tissues have the highest GPER mRNA and protein expressions. The expression is lowest in all db/db. GPER downregulation is associated with promoter hypermethylation and reduced enrichment of H3K4me3 and H3K27ac marks around the GPER promoter. We also observed sex-specific disparities in fecal pellet production patterns of the GPER KO mice compared to WT. We thus, conclude that T2D impairs gut GPER expression, and epigenetic sex-specific mechanisms matter in the downregulation.
Collapse
Affiliation(s)
- Aliyu Muhammad
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, P.M.B. 1044, Zaria, Kaduna State, Nigeria
| | - Juanita C Hixon
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | | | - Jatna I Rivas Zarete
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| | - India Johnson
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Jamial Miller
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Benjamin Adu-Addai
- Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Clayton Yates
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sunila Mahavadi
- Department of Biology, Center for Cancer Research, Tuskegee University, Tuskegee, AL, 36088, USA.
| |
Collapse
|
28
|
Luong RAM, Guan W, Vue FC, Dai J. Literary Identification of Differentially Hydroxymethylated DNA Regions for Type 2 Diabetes Mellitus: A Scoping Minireview. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:177. [PMID: 38397668 PMCID: PMC10887687 DOI: 10.3390/ijerph21020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a public health condition where environmental and genetic factors can intersect through hydroxymethylation. It was unclear which blood DNA regions were hydroxymethylated in human T2DM development. We aimed to identify the regions from the literature as designed in the ongoing Twins Discordant for Incident T2DM Study. A scoping review was performed using Medical Subject Headings (MeSH) and keyword methods to search PubMed for studies published in English and before 1 August 2022, following our registered protocol. The keyword and MeSH methods identified 12 and 3 records separately, and the keyword-identified records included all from the MeSH. Only three case-control studies met the criteria for the full-text review, including one MeSH-identified record. Increased global levels of 5-hydroxymethylated cytosine (5hmC) in T2DM patients versus healthy controls in blood or peripheral blood mononuclear cells were consistently reported (p < 0.05 for all). Among candidate DNA regions related to the human SOCS3, SREBF1, and TXNIP genes, only the SOCS3 gene yielded higher 5hmC levels in T2DM patients with high poly-ADP-ribosylation than participants combined from those with low PARylation and healthy controls (p < 0.05). Hydroxymethylation in the SOCS3-related region of blood DNA is promising to investigate for its mediation in the influences of environment on incident T2DM.
Collapse
Affiliation(s)
- Ryan Anh Minh Luong
- Doctoral Program of Osteopathic Medicine, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (R.A.M.L.); (F.C.V.)
| | - Weihua Guan
- Division of Biostatistics & Health Data Science, University of Minnesota School of Public Health, Minneapolis, MN 55414, USA;
| | - Fue Chee Vue
- Doctoral Program of Osteopathic Medicine, College of Osteopathic Medicine, Des Moines University, West Des Moines, IA 50266, USA; (R.A.M.L.); (F.C.V.)
| | - Jun Dai
- Department of Public Health, College of Health Sciences, Des Moines University, West Des Moines, IA 50266, USA
| |
Collapse
|
29
|
Xu F, Jin J, Guo J, Xu F, Chen J, Liu Q, Song L, Zhang Z, Zhou L, Su J, Xiao C, Zhang Y, Yan M, He Q, Wu D, Chang C, Li X, Wu L. The clinical characteristics, gene mutations and outcomes of myelodysplastic syndromes with diabetes mellitus. J Cancer Res Clin Oncol 2024; 150:71. [PMID: 38305890 PMCID: PMC10837231 DOI: 10.1007/s00432-023-05591-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/22/2023] [Indexed: 02/03/2024]
Abstract
PURPOSE Diabetes mellitus (DM) is the second most common comorbidity in myelodysplastic syndromes (MDS). The purpose of the study was to investigate the clinical characteristics of MDS patients with DM. METHODS A retrospective analysis was performed on the clinical data of 890 MDS patients with or without DM. Clinical data, including genetic changes, overall survival (OS), leukemia-free survival (LFS) and infection, were analyzed. RESULTS Among 890 patients, 184 (20.7%) had DM. TET2 and SF3B1 mutations occurred more frequently in the DM group than those in the non-DM group (p = 0.0092 and p = 0.0004, respectively). Besides, DM was an independent risk factor for infection (HR 2.135 CI 1.451-3.110, p = 0.000) in MDS. Compared to non-DM patients, MDS patients with DM had poor OS and LFS (p = 0.0002 and p = 0.0017, respectively), especially in the lower-risk group. While in multivariate analysis, DM did not retain its prognostic significance and the prognostic significance of infection was maintained (HR 2.488 CI 1.749-3.538, p = 0.000). CONCLUSIONS MDS patients with DM have an inferior prognosis which may due to higher infection incidence, with TET2 and SF3B1 mutations being more frequent in those cases.
Collapse
Affiliation(s)
- Fanhuan Xu
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jiacheng Jin
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Juan Guo
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Feng Xu
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jianan Chen
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qi Liu
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Luxi Song
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zheng Zhang
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Liyu Zhou
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Department of Hematology, Shanghai Jiao Eighth People's Hospital, Shanghai, 200233, China
| | - Jiying Su
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chao Xiao
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yumei Zhang
- Department of Hematology, Shanghai Jiao Eighth People's Hospital, Shanghai, 200233, China
| | - Meng Yan
- Department of Hematology, Shanghai Jiao Eighth People's Hospital, Shanghai, 200233, China
| | - Qi He
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Dong Wu
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chunkang Chang
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Xiao Li
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Lingyun Wu
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Department of Hematology, Shanghai Jiao Eighth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
30
|
Jiang Y, Deng G, Liu C, Tang H, Zheng J, Kong X, Zhao M, Liu Y, Gao P, Li T, Zhao H, Cao Y, Li P, Ma L. Tangshen formula improves diabetic nephropathy in STZ-induced diabetes rats fed with hyper-methionine by regulating the methylation status of kidney. Clin Epigenetics 2024; 16:1. [PMID: 38167534 PMCID: PMC10763145 DOI: 10.1186/s13148-023-01620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The objective of this study was to examine and analyze differential methylation profiles in order to investigate the influence of hyper-methioninemia (HM) on the development of diabetic nephropathy (DN). Male Wistar rats, aged eight weeks and weighing 250-300 g, were randomly assigned into four groups: a control group (Healthy, n = 8), streptozocin-induced rats (STZ group, n = 8), HM + STZ group (n = 8), and the Tangshen Formula (TSF) treatment group (TSF group, n = 8). Blood glucose levels and other metabolic indicators were monitored before treatment and at four-week intervals until 12 weeks. Total DNA was extracted from the aforementioned groups, and DNA methylation landscapes were analyzed via reduced representative bisulfite sequencing. RESULTS Both the STZ group and HM + STZ group exhibited increased blood glucose levels and urinary albumin/creatinine ratios in comparison with the control group. Notably, the HM + STZ group exhibited a markedly elevated urinary albumin/creatinine ratio (411.90 ± 88.86 mg/g) compared to the STZ group (238.41 ± 62.52 mg/g). TSF-treated rats demonstrated substantial reductions in both blood glucose levels and urinary albumin/creatinine ratios in comparison with the HM + STZ group. In-depth analysis of DNA methylation profiles revealed 797 genes with potential therapeutic effects related to TSF, among which approximately 2.3% had been previously reported as homologous genes. CONCLUSION While HM exacerbates DN through altered methylation patterns at specific CpG sites, TSF holds promise as a viable treatment for DN by restoring abnormal methylation levels. The identification of specific genes provides valuable insights into the underlying mechanisms of DN pathogenesis and offers potential therapeutic targets for further investigation.
Collapse
Affiliation(s)
- Yongwei Jiang
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - GuoXiong Deng
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Chengyin Liu
- BioChain (Beijing) Science and Technology Inc., No. 18 Hongda South Road, BDA, Beijing, 100176, China
| | - Han Tang
- BioChain (Beijing) Science and Technology Inc., No. 18 Hongda South Road, BDA, Beijing, 100176, China
| | - Jing Zheng
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Xiaomu Kong
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Meimei Zhao
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Yi Liu
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Peng Gao
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Tianbao Li
- BioChain (Beijing) Science and Technology Inc., No. 18 Hongda South Road, BDA, Beijing, 100176, China
| | - Hailing Zhao
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China
| | - Yongtong Cao
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China.
| | - Ping Li
- Beijing Key Lab Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Science, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China.
| | - Liang Ma
- Clinical Laboratory, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
31
|
Geiger M, Gorica E, Mohammed SA, Mongelli A, Mengozi A, Delfine V, Ruschitzka F, Costantino S, Paneni F. Epigenetic Network in Immunometabolic Disease. Adv Biol (Weinh) 2024; 8:e2300211. [PMID: 37794610 DOI: 10.1002/adbi.202300211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Although a large amount of data consistently shows that genes affect immunometabolic characteristics and outcomes, epigenetic mechanisms are also heavily implicated. Epigenetic changes, including DNA methylation, histone modification, and noncoding RNA, determine gene activity by altering the accessibility of chromatin to transcription factors. Various factors influence these alterations, including genetics, lifestyle, and environmental cues. Moreover, acquired epigenetic signals can be transmitted across generations, thus contributing to early disease traits in the offspring. A closer investigation is critical in this aspect as it can help to understand the underlying molecular mechanisms further and gain insights into potential therapeutic targets for preventing and treating diseases arising from immuno-metabolic dysregulation. In this review, the role of chromatin alterations in the transcriptional modulation of genes involved in insulin resistance, systemic inflammation, macrophage polarization, endothelial dysfunction, metabolic cardiomyopathy, and nonalcoholic fatty liver disease (NAFLD), is discussed. An overview of emerging chromatin-modifying drugs and the importance of the individual epigenetic profile for personalized therapeutic approaches in patients with immuno-metabolic disorders is also presented.
Collapse
Affiliation(s)
- Martin Geiger
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessandro Mengozi
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- Department of Research and Education, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| |
Collapse
|
32
|
Amine Ikhanjal M, Ali Elouarid M, Zouine C, El Alami H, Errafii K, Ghazal H, Alidrissi N, Bakkali F, Benmoussa A, Hamdi S. FTO gene variants (rs9939609, rs8050136 and rs17817449) and type 2 diabetes mellitus risk: A Meta-Analysis. Gene 2023; 887:147791. [PMID: 37696421 DOI: 10.1016/j.gene.2023.147791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/31/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
BACKGROUND AND AIMS There is tremendous increase in type 2 diabetes mellitus (T2DM) worldwide. The impact of FTO gene polymorphisms on the risk of T2DM is not yet clear because of the controversial results of studies. This meta-analysis aimed to better clarify the association between three FTO gene polymorphisms SNPs (rs9939609, rs8050136 and rs17817449) and T2DM in a larger combined population worldwide. MATERIAL AND METHODS A comprehensive search on the PubMed, Science Direct, and Web of Science databases was conducted to identify investigations in relationship between different FTO gene polymorphisms (rs9939609, rs8050136 and rs17817449) and T2DM globally. Published papers from January 2007 to May 2023 were collected. Inclusion criteria are limited to human case-control studies published in English and peer-reviewed, which provided data on the genotype distributions of FTO gene polymorphisms and T2DM risk. Odds ratios (OR) and 95% confidence intervals (CI) were calculated to express the results of the meta-analysis. Potential sources of bias and heterogeneity using Egger's regression analysis were also assessed. RESULTS Of 234695 identified articles, forty-eight studies were selected including 36,051 patients with T2DM and 51,266 control subjects. Overall, we found a significant increased risk of T2DM susceptibility and rs9939609 FTO gene polymorphism in the Allele contrast (A vs. T: OR = 1,30, 95% CI = 1.14; 1.48, P < 0,05, I2 = 0,94), Recessive model (AA vs. AT + TT: OR = 1,54, 95% CI = 1.19; 2.00, P < 0,05, I2 = 0,94), Dominant model (AA + AT vs. TT: OR = 1,26, 95% CI = 1.10; 1.45, P < 0,05, I2 = 0,89), homozygote model (AA vs. TT: OR = 1,60, 95% CI = 1.26; 2.03, P < 0,05, I2 = 0,90), and heterozygote model (AA vs. AT: OR = 1,43, 95% CI = 1.09; 1.88, P = 0,008, I2 = 0,93). we also found a significantly increased risk of T2DM susceptibility and rs8050136 FTO gene polymorphism under all models. For rs17817449 we did not find any association between with T2DM. CONCLUSION The present meta-analysis confirms that rs9939609 and rs8050136 in the FTO gene are significantly associated with T2DM, while rs17817449 does not show any association.
Collapse
Affiliation(s)
- Mohammed Amine Ikhanjal
- Environmental Health Laboratory, Institut Pasteur du Maroc, Morocco; University of Mohamed VI of Sciences and Health, Morocco.
| | - Mohammed Ali Elouarid
- Environmental Health Laboratory, Institut Pasteur du Maroc, Morocco; University of Mohamed VI of Sciences and Health, Morocco.
| | - Chaimae Zouine
- Environmental Health Laboratory, Institut Pasteur du Maroc, Morocco; University of Mohamed VI of Sciences and Health, Morocco.
| | - Houda El Alami
- Environmental Health Laboratory, Institut Pasteur du Maroc, Morocco.
| | - Khaoula Errafii
- African Genomic Center (AGC), University Mohamed VI Polytechnic, Bengurir, Morocco.
| | - Hassan Ghazal
- Laboratory of Genomics, Bioinformatics and Digital Health, School of Medicine, Mohammed VI University of Science and Health, Casablanca, Morocco;s Royal Institute for Management Training, Rabat, Morocco.
| | - Najib Alidrissi
- Department of Surgery and Laboratory of Genomics, Bioinformatics and Digital Health, School of Medicine, Mohammed VI University of Health Sciences, Casablanca, Morocco; Hospital Cheikh Khalifa, Casablanca, Morocco.
| | - Fadil Bakkali
- University of Mohamed VI of Sciences and Health, Morocco; Laboratory of toxicology, toxicogenomics and ecotoxicology, University of Mohamed VI of Sciences and Health, Morocco.
| | - Adnane Benmoussa
- University of Mohamed VI of Sciences and Health, Morocco; Laboratory of toxicology, toxicogenomics and ecotoxicology, University of Mohamed VI of Sciences and Health, Morocco.
| | - Salsabil Hamdi
- Environmental Health Laboratory, Institut Pasteur du Maroc, Morocco.
| |
Collapse
|
33
|
Keshawarz A, Joehanes R, Ma J, Lee GY, Costeira R, Tsai PC, Masachs OM, Bell JT, Wilson R, Thorand B, Winkelmann J, Peters A, Linseisen J, Waldenberger M, Lehtimäki T, Mishra PP, Kähönen M, Raitakari O, Helminen M, Wang CA, Melton PE, Huang RC, Pennell CE, O’Sullivan TA, Ochoa-Rosales C, Voortman T, van Meurs JB, Young KL, Graff M, Wang Y, Kiel DP, Smith CE, Jacques PF, Levy D. Dietary and supplemental intake of vitamins C and E is associated with altered DNA methylation in an epigenome-wide association study meta-analysis. Epigenetics 2023; 18:2211361. [PMID: 37233989 PMCID: PMC10228397 DOI: 10.1080/15592294.2023.2211361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 04/28/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Dietary intake of antioxidants such as vitamins C and E protect against oxidative stress, and may also be associated with altered DNA methylation patterns. METHODS We meta-analysed epigenome-wide association study (EWAS) results from 11,866 participants across eight population-based cohorts to evaluate the association between self-reported dietary and supplemental intake of vitamins C and E with DNA methylation. EWAS were adjusted for age, sex, BMI, caloric intake, blood cell type proportion, smoking status, alcohol consumption, and technical covariates. Significant results of the meta-analysis were subsequently evaluated in gene set enrichment analysis (GSEA) and expression quantitative trait methylation (eQTM) analysis. RESULTS In meta-analysis, methylation at 4,656 CpG sites was significantly associated with vitamin C intake at FDR ≤ 0.05. The most significant CpG sites associated with vitamin C (at FDR ≤ 0.01) were enriched for pathways associated with systems development and cell signalling in GSEA, and were associated with downstream expression of genes enriched in the immune response in eQTM analysis. Furthermore, methylation at 160 CpG sites was significantly associated with vitamin E intake at FDR ≤ 0.05, but GSEA and eQTM analysis of the top most significant CpG sites associated with vitamin E did not identify significant enrichment of any biological pathways investigated. CONCLUSIONS We identified significant associations of many CpG sites with vitamin C and E intake, and our results suggest that vitamin C intake may be associated with systems development and the immune response.
Collapse
Affiliation(s)
| | - Roby Joehanes
- Framingham Heart Study, Framingham, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jiantao Ma
- Framingham Heart Study, Framingham, Framingham, MA, USA
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Gha Young Lee
- Framingham Heart Study, Framingham, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ricardo Costeira
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Olatz M. Masachs
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jordana T. Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Rory Wilson
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, Neuherberg, Germany
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, München, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
- Chair of Neurogenetics, School of Medicine, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, München, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Chair of Epidemiology, Medical Faculty, Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), München Heart Alliance, Munich, Germany
| | - Jakob Linseisen
- Chair of Epidemiology, University Augsburg at University Hospital Augsburg, Augsburg, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), München Heart Alliance, Munich, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Pashupati P. Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Olli Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Mika Helminen
- Tays Research Services, Tampere University Hospital, Tampere, Finland
- Faculty of Social Sciences, Health Sciences, Tampere University, Tampere, Finland
| | - Carol A. Wang
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Phillip E. Melton
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- School of Population and Global Health, University of Western Australia, Perth, Australia
| | - Rae-Chi Huang
- Nutrition & Health Innovation Research Institute, Edith Cowan University, Perth, Australia
| | - Craig E. Pennell
- Faculty of Social Sciences, Health Sciences, Tampere University, Tampere, Finland
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | | | - Carolina Ochoa-Rosales
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
- Centro de Vida Saludable, Universidad de Concepción, Concepción, Chile
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Joyce B.J. van Meurs
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Kristin L. Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Hebrew Senior Life, Chapel Hill, North Carolina, USA
| | - Misa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Hebrew Senior Life, Chapel Hill, North Carolina, USA
| | - Yujie Wang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Hebrew Senior Life, Chapel Hill, North Carolina, USA
| | - Douglas P. Kiel
- Department of Medicine, Beth Israel Deaconess Medical Center, Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Caren E. Smith
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Paul F. Jacques
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Daniel Levy
- Framingham Heart Study, Framingham, Framingham, MA, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
34
|
Deng JY, Wu XQ, He WJ, Liao X, Tang M, Nie XQ. Targeting DNA methylation and demethylation in diabetic foot ulcers. J Adv Res 2023; 54:119-131. [PMID: 36706989 PMCID: PMC10703625 DOI: 10.1016/j.jare.2023.01.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Poor wound healing is a significant complication of diabetes, which is commonly caused by neuropathy, trauma, deformities, plantar hypertension and peripheral arterial disease. Diabetic foot ulcers (DFU) are difficult to heal, which makes patients susceptible to infections and can ultimately conduce to limb amputation or even death in severe cases. An increasing number of studies have found that epigenetic alterations are strongly associated with poor wound healing in diabetes. AIM OF REVIEW This work provides significant insights into the development of therapeutics for improving chronic diabetic wound healing, particularly by targeting and regulating DNA methylation and demethylation in DFU. Key scientific concepts of review: DNA methylation and demethylation play an important part in diabetic wound healing, via regulating corresponding signaling pathways in different breeds of cells, including macrophages, vascular endothelial cells and keratinocytes. In this review, we describe the four main phases of wound healing and their abnormality in diabetic patients. Furthermore, we provided an in-depth summary and discussion on how DNA methylation and demethylation regulate diabetic wound healing in different types of cells; and gave a brief summary on recent advances in applying cellular reprogramming techniques for improving diabetic wound healing.
Collapse
Affiliation(s)
- Jun-Yu Deng
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563006, China; Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Xing-Qian Wu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Wen-Jie He
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Xin Liao
- Affiliated Hospital of Zunyi Medical University, Zunyi 563006, China
| | - Ming Tang
- Queensland University of Technology (QUT), School of Biomedical Sciences, Centre for Genomics and Personalized Health at the Translational Research Institute (TRI), Brisbane, QLD 4102, Australia.
| | - Xu-Qiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Zunyi 563006, China; Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Queensland University of Technology (QUT), School of Biomedical Sciences, Centre for Genomics and Personalized Health at the Translational Research Institute (TRI), Brisbane, QLD 4102, Australia.
| |
Collapse
|
35
|
Tatsch JM, Furman DP, Nobre RM, Wurzer KM, da Silva LC, Picheth GF, Ramos EA, Acco A, Klassen G. Dulaglutide as a demethylating agent to improve the outcome of breast cancer. Epigenomics 2023; 15:1309-1322. [PMID: 38174426 DOI: 10.2217/epi-2023-0332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
Abstract
Background: Dulaglutide emerged as a promising therapeutic option for diabetes mellitus Type 2 (DM2). Aims: Owing to epigenetic similarities between the pathophysiology of DM2 and breast cancer (BC), we investigated the antitumor effect of dulaglutide. Materials & methods: To investigate the effect of dulaglutide, we analyzed the expression of methylated gene promoter regions in BC (ESR1, CDH1 and ADAM33). Results: Dulaglutide increased the expression of ESR1, CDH1 and ADAM33 up to fourfold in the MDA-MB-231 lineage by demethylating the gene promoter regions. This effect was translated to in vivo antitumoral activity and revealed significant tumor inhibition by combining the half-dose of methotrexate with dulaglutide. Conclusion: This therapy may mitigate the severe side effects commonly associated with chemotherapy.
Collapse
Affiliation(s)
- Júlia M Tatsch
- Department of Basic Pathology, Laboratory of Epigenetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Diana P Furman
- Department of Basic Pathology, Laboratory of Epigenetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Rodrigo Mb Nobre
- Department of Basic Pathology, Laboratory of Epigenetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Karin M Wurzer
- Department of Basic Pathology, Laboratory of Epigenetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Liziane Cm da Silva
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Guilherme F Picheth
- Department of Biochemistry Federal University of Paraná, Curitiba, PR, Brazil
| | - Edneia As Ramos
- Department of Basic Pathology, Laboratory of Epigenetics, Federal University of Paraná, Curitiba, PR, Brazil
| | - Alexandra Acco
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Giseli Klassen
- Department of Basic Pathology, Laboratory of Epigenetics, Federal University of Paraná, Curitiba, PR, Brazil
| |
Collapse
|
36
|
Ustianowski Ł, Udzik J, Szostak J, Gorący A, Ustianowska K, Pawlik A. Genetic and Epigenetic Factors in Gestational Diabetes Mellitus Pathology. Int J Mol Sci 2023; 24:16619. [PMID: 38068941 PMCID: PMC10706782 DOI: 10.3390/ijms242316619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Gestational diabetes (GDM) is the carbohydrate intolerance occurring during pregnancy. The risk factors of GDM include obesity, advanced maternal age, polycystic ovary syndrome, multigravidity, a sedentary lifestyle, and pre-existing hypertension. Additionally, complex genetic and epigenetic processes are also believed to play a crucial role in the development of GDM. In this narrative review, we discuss the role of genetic and epigenetic factors in gestational diabetes mellitus pathogenesis.
Collapse
Affiliation(s)
- Łukasz Ustianowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (Ł.U.); (J.U.); (K.U.)
| | - Jakub Udzik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (Ł.U.); (J.U.); (K.U.)
- Department of Cardiac Surgery, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Joanna Szostak
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Anna Gorący
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Klaudia Ustianowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (Ł.U.); (J.U.); (K.U.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (Ł.U.); (J.U.); (K.U.)
| |
Collapse
|
37
|
Thongsroy J, Mutirangura A. The inverse association between DNA gaps and HbA1c levels in type 2 diabetes mellitus. Sci Rep 2023; 13:18987. [PMID: 37923892 PMCID: PMC10624909 DOI: 10.1038/s41598-023-46431-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023] Open
Abstract
Naturally occurring DNA gaps have been observed in eukaryotic DNA, including DNA in nondividing cells. These DNA gaps are found less frequently in chronologically aging yeast, chemically induced senescence cells, naturally aged rats, D-galactose-induced aging model rats, and older people. These gaps function to protect DNA from damage, so we named them youth-associated genomic stabilization DNA gaps (youth-DNA-gaps). Type 2 diabetes mellitus (type 2 DM) is characterized by an early aging phenotype. Here, we explored the correlation between youth-DNA-gaps and the severity of type 2 DM. Here, we investigated youth-DNA-gaps in white blood cells from normal controls, pre-DM, and type 2 DM patients. We found significantly decreased youth-DNA-gap numbers in the type 2 DM patients compared to normal controls (P = 0.0377, P = 0.0018 adjusted age). In the type 2 DM group, youth-DNA-gaps correlate directly with HbA1c levels. (r = - 0.3027, P = 0.0023). Decreased youth-DNA-gap numbers were observed in patients with type 2 DM and associated with increased HbA1c levels. Therefore, the decrease in youth-DNA-gaps is associated with the molecular pathogenesis of high blood glucose levels. Furthermore, youth-DNA-gap number is another marker that could be used to determine the severity of type 2 DM.
Collapse
Affiliation(s)
- Jirapan Thongsroy
- School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
| | - Apiwat Mutirangura
- Center for Excellence in Molecular Genetics of Cancer and Human Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
38
|
Camacho P, Ribeiro E, Pereira B, Varandas T, Nascimento J, Henriques J, Dutra-Medeiros M, Delgadinho M, Oliveira K, Silva C, Brito M. DNA methyltransferase expression (DNMT1, DNMT3a and DNMT3b) as a potential biomarker for anti-VEGF diabetic macular edema response. Eur J Ophthalmol 2023; 33:2267-2274. [PMID: 37082811 PMCID: PMC10590013 DOI: 10.1177/11206721231171623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
PURPOSE DNA methylation is involved in Diabetic Retinopathy progression showing a metabolic memory mechanism. However, the association of DNA methyltransferase with diabetic macular edema is still unknown. We aimed to describe the differences in DNA methyltransferase gene expression in patients with different diabetic macular edema responses. METHODS A total of 27 diabetic patients, aged 59-90 years, were prospectively enrolled in this cross-sectional study. The participants were classified into control group (CG, n = 11), diabetic macular edema responders (rDME, n = 9) and non-responder diabetic macular edema (nrDME, n = 7) after anti-vascular endothelial growth factor (anti-VEGF) treatment. Only cases with a complete ophthalmological examination, digital 133° color fundus, and SD-OCT assessments were used. After RNA extraction and first-strand cDNA synthesis, quantitative real-time PCR was performed with specific primers on the CFX Connect™ Real-Time PCR Detection System to assess differential transcriptional expression patterns. RESULTS The DNMT1 gene showed a positive correlation (r = 0.617; p = 0.043) with Best Corrected Visual Acuity (BCVA) in CG, a positive correlation (r = 0.917; p = 0.010) with HbA1c in nrDME and a negative correlation (r = -0.659; p = 0.049) with GCL-IPL thickness in rDME. DNMT3A gene showed a positive correlation (r = -0.890; p = 0.001) with Sub-foveal Choroidal thickness in rDME whereas DNMT3b gene showed a negative correlation (r = -0.815; p = 0.007) with HbA1c and RNFL (r = -0.664; p = 0.026) in CG. CONCLUSIONS Patients with similar metabolic profile risk factors showed associated DNA methyltransferase transcriptional expression patterns differences fitting with the anti-VEGF diabetic macular edema response. Further studies are needed to clarify if these results (1) reflect disease evolution, (2) translate the therapeutic impact, (3) or can help to predict the therapeutic resistance profile.
Collapse
Affiliation(s)
- Pedro Camacho
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
- Ophtalmology Institute Dr. Gama Pinto, Lisbon, Portugal
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Edna Ribeiro
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Bruno Pereira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Retina Institute of Lisbon, Lisbon, Portugal
| | | | - João Nascimento
- Retina Institute of Lisbon, Lisbon, Portugal
- Beatriz Ângelo Hospital, Lisbon, Portugal
| | | | - Marco Dutra-Medeiros
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
- Central Lisbon Hospital Center, Lisbon, Portugal
| | - Mariana Delgadinho
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Ketlyn Oliveira
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Carina Silva
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Miguel Brito
- H&TRC- Health & Technology Research Center, ESTeSL- Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal
| |
Collapse
|
39
|
Sarnowski C, Huan T, Ma Y, Joehanes R, Beiser A, DeCarli CS, Heard-Costa NL, Levy D, Lin H, Liu CT, Liu C, Meigs JB, Satizabal CL, Florez JC, Hivert MF, Dupuis J, De Jager PL, Bennett DA, Seshadri S, Morrison AC. Multi-tissue epigenetic analysis identifies distinct associations underlying insulin resistance and Alzheimer's disease at CPT1A locus. Clin Epigenetics 2023; 15:173. [PMID: 37891690 PMCID: PMC10612362 DOI: 10.1186/s13148-023-01589-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Insulin resistance (IR) is a major risk factor for Alzheimer's disease (AD) dementia. The mechanisms by which IR predisposes to AD are not well-understood. Epigenetic studies may help identify molecular signatures of IR associated with AD, thus improving our understanding of the biological and regulatory mechanisms linking IR and AD. METHODS We conducted an epigenome-wide association study of IR, quantified using the homeostatic model assessment of IR (HOMA-IR) and adjusted for body mass index, in 3,167 participants from the Framingham Heart Study (FHS) without type 2 diabetes at the time of blood draw used for methylation measurement. We identified DNA methylation markers associated with IR at the genome-wide level accounting for multiple testing (P < 1.1 × 10-7) and evaluated their association with neurological traits in participants from the FHS (N = 3040) and the Religious Orders Study/Memory and Aging Project (ROSMAP, N = 707). DNA methylation profiles were measured in blood (FHS) or dorsolateral prefrontal cortex (ROSMAP) using the Illumina HumanMethylation450 BeadChip. Linear regressions (ROSMAP) or mixed-effects models accounting for familial relatedness (FHS) adjusted for age, sex, cohort, self-reported race, batch, and cell type proportions were used to assess associations between DNA methylation and neurological traits accounting for multiple testing. RESULTS We confirmed the strong association of blood DNA methylation with IR at three loci (cg17901584-DHCR24, cg17058475-CPT1A, cg00574958-CPT1A, and cg06500161-ABCG1). In FHS, higher levels of blood DNA methylation at cg00574958 and cg17058475 were both associated with lower IR (P = 2.4 × 10-11 and P = 9.0 × 10-8), larger total brain volumes (P = 0.03 and P = 9.7 × 10-4), and smaller log lateral ventricular volumes (P = 0.07 and P = 0.03). In ROSMAP, higher levels of brain DNA methylation at the same two CPT1A markers were associated with greater risk of cognitive impairment (P = 0.005 and P = 0.02) and higher AD-related indices (CERAD score: P = 5 × 10-4 and 0.001; Braak stage: P = 0.004 and P = 0.01). CONCLUSIONS Our results suggest potentially distinct epigenetic regulatory mechanisms between peripheral blood and dorsolateral prefrontal cortex tissues underlying IR and AD at CPT1A locus.
Collapse
Affiliation(s)
- Chloé Sarnowski
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Tianxiao Huan
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD, USA
| | - Yiyi Ma
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung and Blood Institutes of Health, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Alexa Beiser
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | | | - Nancy L Heard-Costa
- The Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Daniel Levy
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Chunyu Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - James B Meigs
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Claudia L Satizabal
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jose C Florez
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine and Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Harvard University, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Josée Dupuis
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
- Department of Epidemiology, Biostatistics and Occupational Health, School of Population and Global Health, McGill University, Montreal, Canada
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sudha Seshadri
- The Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
40
|
Butnariu LI, Gorduza EV, Țarcă E, Pânzaru MC, Popa S, Stoleriu S, Lupu VV, Lupu A, Cojocaru E, Trandafir LM, Moisă ȘM, Florea A, Stătescu L, Bădescu MC. Current Data and New Insights into the Genetic Factors of Atherogenic Dyslipidemia Associated with Metabolic Syndrome. Diagnostics (Basel) 2023; 13:2348. [PMID: 37510094 PMCID: PMC10378477 DOI: 10.3390/diagnostics13142348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Atherogenic dyslipidemia plays a critical role in the development of metabolic syndrome (MetS), being one of its major components, along with central obesity, insulin resistance, and hypertension. In recent years, the development of molecular genetics techniques and extended analysis at the genome or exome level has led to important progress in the identification of genetic factors (heritability) involved in lipid metabolism disorders associated with MetS. In this review, we have proposed to present the current knowledge related to the genetic etiology of atherogenic dyslipidemia, but also possible challenges for future studies. Data from the literature provided by candidate gene-based association studies or extended studies, such as genome-wide association studies (GWAS) and whole exome sequencing (WES,) have revealed that atherogenic dyslipidemia presents a marked genetic heterogeneity (monogenic or complex, multifactorial). Despite sustained efforts, many of the genetic factors still remain unidentified (missing heritability). In the future, the identification of new genes and the molecular mechanisms by which they intervene in lipid disorders will allow the development of innovative therapies that act on specific targets. In addition, the use of polygenic risk scores (PRS) or specific biomarkers to identify individuals at increased risk of atherogenic dyslipidemia and/or other components of MetS will allow effective preventive measures and personalized therapy.
Collapse
Affiliation(s)
- Lăcramioara Ionela Butnariu
- Department of Medical Genetics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Eusebiu Vlad Gorduza
- Department of Medical Genetics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Elena Țarcă
- Department of Surgery II-Pediatric Surgery, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Monica-Cristina Pânzaru
- Department of Medical Genetics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Setalia Popa
- Department of Medical Genetics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Simona Stoleriu
- Odontology-Periodontology, Fixed Prosthesis Department, Faculty of Dental Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Vasile Valeriu Lupu
- Department of Pediatrics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ancuta Lupu
- Department of Pediatrics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Elena Cojocaru
- Department of Morphofunctional Sciences I, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laura Mihaela Trandafir
- Department of Pediatrics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ștefana Maria Moisă
- Department of Pediatrics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Andreea Florea
- Department of Medical Genetics, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Laura Stătescu
- Medical III Department, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Minerva Codruța Bădescu
- III Internal Medicine Clinic, "St. Spiridon" County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
41
|
Abubakar B, Usman D, Sanusi KO, Azmi NH, Imam MU. Preventive Epigenetic Mechanisms of Functional Foods for Type 2 Diabetes. DIABETOLOGY 2023; 4:259-277. [DOI: 10.3390/diabetology4030023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Type 2 diabetes (T2D) is a growing global health problem that requires new and effective prevention and management strategies. Recent research has highlighted the role of epigenetic changes in the development and progression of T2D, and the potential of functional foods as a complementary therapy for the disease. This review aims to provide an overview of the current state of knowledge on the preventive epigenetic mechanisms of functional foods in T2D. We provide background information on T2D and its current treatment approaches, an explanation of the concept of epigenetics, and an overview of the different functional foods with demonstrated preventive epigenetic effects in T2D. We also discuss the epigenetic mechanisms by which these functional foods prevent or manage T2D, and the studies that have investigated their preventive epigenetic effects. In addition, we revisit works on the beneficial influence of functional foods against the programming and complications of parentally-triggered offspring diabetes. We also suggest, albeit based on scarce data, that epigenetic inheritance mechanistically mediates the impacts of functional nutrition against the metabolic risk of diabetes in offspring. Finally, our review highlights the importance of considering the preventive epigenetic mechanisms of functional foods as a potential avenue for the development of new prevention and management strategies for T2D.
Collapse
Affiliation(s)
- Bilyaminu Abubakar
- Department of Pharmacology and Toxicology, Usmanu Danfodiyo University, Sokoto P.M.B. 2346, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto P.M.B. 2346, Nigeria
| | - Dawoud Usman
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto P.M.B. 2346, Nigeria
- Department of Physiology, Usmanu Danfodiyo University, Sokoto P.M.B. 2346, Nigeria
| | - Kamaldeen Olalekan Sanusi
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto P.M.B. 2346, Nigeria
- Department of Physiology, Usmanu Danfodiyo University, Sokoto P.M.B. 2346, Nigeria
| | - Nur Hanisah Azmi
- Faculty of Food Science and Nutrition, Universiti Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Mustapha Umar Imam
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto P.M.B. 2346, Nigeria
- Department of Medical Biochemistry, Usmanu Danfodiyo University, Sokoto P.M.B. 2346, Nigeria
| |
Collapse
|
42
|
Ray D, Loomis SJ, Venkataraghavan S, Tin A, Yu B, Chatterjee N, Selvin E, Duggal P. Characterizing common and rare variations in non-traditional glycemic biomarkers using multivariate approaches on multi-ancestry ARIC study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.13.23289200. [PMID: 37398180 PMCID: PMC10312851 DOI: 10.1101/2023.06.13.23289200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Glycated hemoglobin, fasting glucose, glycated albumin, and fructosamine are biomarkers that reflect different aspects of the glycemic process. Genetic studies of these glycemic biomarkers can shed light on unknown aspects of type 2 diabetes genetics and biology. While there exists several GWAS of glycated hemoglobin and fasting glucose, very few GWAS have focused on glycated albumin or fructosamine. We performed a multi-phenotype GWAS of glycated albumin and fructosamine from 7,395 White and 2,016 Black participants in the Atherosclerosis Risk in Communities (ARIC) study on the common variants from genotyped/imputed data. We found 2 genome-wide significant loci, one mapping to known type 2 diabetes gene (ARAP1/STARD10, p = 2.8 × 10-8) and another mapping to a novel gene (UGT1A, p = 1.4 × 10-8) using multi-omics gene mapping strategies in diabetes-relevant tissues. We identified additional loci that were ancestry-specific (e.g., PRKCA from African ancestry individuals, p = 1.7 × 10-8) and sex-specific (TEX29 locus in males only, p = 3.0 × 10-8). Further, we implemented multi-phenotype gene-burden tests on whole-exome sequence data from 6,590 White and 2,309 Black ARIC participants. Eleven genes across different rare variant aggregation strategies were exome-wide significant only in multi-ancestry analysis. Four out of 11 genes had notable enrichment of rare predicted loss of function variants in African ancestry participants despite smaller sample size. Overall, 8 out of 15 loci/genes were implicated to influence these biomarkers via glycemic pathways. This study illustrates improved locus discovery and potential effector gene discovery by leveraging joint patterns of related biomarkers across entire allele frequency spectrum in multi-ancestry analyses. Most of the loci/genes we identified have not been previously implicated in studies of type 2 diabetes, and future investigation of the loci/genes potentially acting through glycemic pathways may help us better understand risk of developing type 2 diabetes.
Collapse
Affiliation(s)
- Debashree Ray
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | | | - Sowmya Venkataraghavan
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| | - Adrienne Tin
- School of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Bing Yu
- Department of Epidemiology, UTHealth School of Public Health, Houston, TX
| | - Nilanjan Chatterjee
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Elizabeth Selvin
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
- Welch Center for Prevention, Epidemiology, & Clinical Research, Johns Hopkins University, Baltimore, MD
| | - Priya Duggal
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD
| |
Collapse
|
43
|
Berlanga-Acosta J, Garcia-Ojalvo A, Guillen-Nieto G, Ayala-Avila M. Endogenous Biological Drivers in Diabetic Lower Limb Wounds Recurrence: Hypothetical Reflections. Int J Mol Sci 2023; 24:10170. [PMID: 37373317 DOI: 10.3390/ijms241210170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
An impaired healing response underlies diabetic foot wound chronicity, frequently translating to amputation, disability, and mortality. Diabetics suffer from underappreciated episodes of post-epithelization ulcer recurrence. Recurrence epidemiological data are alarmingly high, so the ulcer is considered in "remission" and not healed from the time it remains epithelialized. Recurrence may result from the combined effects of behavioral and endogenous biological factors. Although the damaging role of behavioral, clinical predisposing factors is undebatable, it still remains elusive in the identification of endogenous biological culprits that may prime the residual scar tissue for recurrence. Furthermore, the event of ulcer recurrence still waits for the identification of a molecular predictor. We propose that ulcer recurrence is deeply impinged by chronic hyperglycemia and its downstream biological effectors, which originate epigenetic drivers that enforce abnormal pathologic phenotypes to dermal fibroblasts and keratinocytes as memory cells. Hyperglycemia-derived cytotoxic reactants accumulate and modify dermal proteins, reduce scar tissue mechanical tolerance, and disrupt fibroblast-secretory activity. Accordingly, the combination of epigenetic and local and systemic cytotoxic signalers induce the onset of "at-risk phenotypes" such as premature skin cell aging, dysmetabolism, inflammatory, pro-degradative, and oxidative programs that may ultimately converge to scar cell demise. Post-epithelialization recurrence rate data are missing in clinical studies of reputed ulcer healing therapies during follow-up periods. Intra-ulcer infiltration of epidermal growth factor exhibits the most consistent remission data with the lowest recurrences during 12-month follow-up. Recurrence data should be regarded as a valuable clinical endpoint during the investigational period for each emergent healing candidate.
Collapse
Affiliation(s)
- Jorge Berlanga-Acosta
- Tissue Repair, Wound Healing and Cytoprotection Research Group, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, Havana 10600, Cuba
| | - Ariana Garcia-Ojalvo
- Tissue Repair, Wound Healing and Cytoprotection Research Group, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, Havana 10600, Cuba
| | - Gerardo Guillen-Nieto
- Tissue Repair, Wound Healing and Cytoprotection Research Group, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, Havana 10600, Cuba
| | - Marta Ayala-Avila
- Tissue Repair, Wound Healing and Cytoprotection Research Group, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology, Playa, Havana 10600, Cuba
| |
Collapse
|
44
|
Frankowski R, Kobierecki M, Wittczak A, Różycka-Kosmalska M, Pietras T, Sipowicz K, Kosmalski M. Type 2 Diabetes Mellitus, Non-Alcoholic Fatty Liver Disease, and Metabolic Repercussions: The Vicious Cycle and Its Interplay with Inflammation. Int J Mol Sci 2023; 24:ijms24119677. [PMID: 37298632 DOI: 10.3390/ijms24119677] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
The prevalence of metabolic-related disorders, such as non-alcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (DM2), has been increasing. Therefore, developing improved methods for the prevention, treatment, and detection of these two conditions is also necessary. In this study, our primary focus was on examining the role of chronic inflammation as a potential link in the pathogenesis of these diseases and their interconnections. A comprehensive search of the PubMed database using keywords such as "non-alcoholic fatty liver disease", "type 2 diabetes mellitus", "chronic inflammation", "pathogenesis", and "progression" yielded 177 relevant papers for our analysis. The findings of our study revealed intricate relationships between the pathogenesis of NAFLD and DM2, emphasizing the crucial role of inflammatory processes. These connections involve various molecular functions, including altered signaling pathways, patterns of gene methylation, the expression of related peptides, and up- and downregulation of several genes. Our study is a foundational platform for future research into the intricate relationship between NAFLD and DM2, allowing for a better understanding of the underlying mechanisms and the potential for introducing new treatment standards.
Collapse
Affiliation(s)
- Rafał Frankowski
- Students' Research Club, Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| | - Mateusz Kobierecki
- Students' Research Club, Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| | - Andrzej Wittczak
- Students' Research Club, Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| | | | - Tadeusz Pietras
- Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| | - Kasper Sipowicz
- Department of Interdisciplinary Disability Studies, The Maria Grzegorzewska University in Warsaw, 02-353 Warsaw, Poland
| | - Marcin Kosmalski
- Department of Clinical Pharmacology, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|
45
|
Padovani-Claudio DA, Ramos CJ, Capozzi ME, Penn JS. Elucidating glial responses to products of diabetes-associated systemic dyshomeostasis. Prog Retin Eye Res 2023; 94:101151. [PMID: 37028118 PMCID: PMC10683564 DOI: 10.1016/j.preteyeres.2022.101151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 04/08/2023]
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness in working age adults. DR has non-proliferative stages, characterized in part by retinal neuroinflammation and ischemia, and proliferative stages, characterized by retinal angiogenesis. Several systemic factors, including poor glycemic control, hypertension, and hyperlipidemia, increase the risk of DR progression to vision-threatening stages. Identification of cellular or molecular targets in early DR events could allow more prompt interventions pre-empting DR progression to vision-threatening stages. Glia mediate homeostasis and repair. They contribute to immune surveillance and defense, cytokine and growth factor production and secretion, ion and neurotransmitter balance, neuroprotection, and, potentially, regeneration. Therefore, it is likely that glia orchestrate events throughout the development and progression of retinopathy. Understanding glial responses to products of diabetes-associated systemic dyshomeostasis may reveal novel insights into the pathophysiology of DR and guide the development of novel therapies for this potentially blinding condition. In this article, first, we review normal glial functions and their putative roles in the development of DR. We then describe glial transcriptome alterations in response to systemic circulating factors that are upregulated in patients with diabetes and diabetes-related comorbidities; namely glucose in hyperglycemia, angiotensin II in hypertension, and the free fatty acid palmitic acid in hyperlipidemia. Finally, we discuss potential benefits and challenges associated with studying glia as targets of DR therapeutic interventions. In vitro stimulation of glia with glucose, angiotensin II and palmitic acid suggests that: 1) astrocytes may be more responsive than other glia to these products of systemic dyshomeostasis; 2) the effects of hyperglycemia on glia are likely to be largely osmotic; 3) fatty acid accumulation may compound DR pathophysiology by promoting predominantly proinflammatory and proangiogenic transcriptional alterations of macro and microglia; and 4) cell-targeted therapies may offer safer and more effective avenues for DR treatment as they may circumvent the complication of pleiotropism in retinal cell responses. Although several molecules previously implicated in DR pathophysiology are validated in this review, some less explored molecules emerge as potential therapeutic targets. Whereas much is known regarding glial cell activation, future studies characterizing the role of glia in DR and how their activation is regulated and sustained (independently or as part of retinal cell networks) may help elucidate mechanisms of DR pathogenesis and identify novel drug targets for this blinding disease.
Collapse
Affiliation(s)
- Dolly Ann Padovani-Claudio
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3321A Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Carla J Ramos
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, AA1324 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| | - Megan E Capozzi
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 North Duke Street, Durham, NC, 27701, USA.
| | - John S Penn
- Department of Ophthalmology and Visual Sciences, Vanderbilt University School of Medicine, B3307 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232-0011, USA.
| |
Collapse
|
46
|
Salah Hasan N, Gamal El Dine H, Ahmed Kamel S, Hamed M, Youssef RN, Mahmoud Hassan E, Ibrahim Musa N, Ali A, Awadallah E. Association of Genetic and Epigenetic changes of Insulin Like Growth Factor Binding Protein-1 in Egyptian Patients with Type 2 Diabetes Mellitus. Diabetes Res Clin Pract 2023; 200:110677. [PMID: 37088243 DOI: 10.1016/j.diabres.2023.110677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 03/20/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND Diabetes is one of the global health threat. Type 2 Diabetes mellitus (T2DM) is associated with life-threatening complications. This work, aimed to study the association between T2DM and IGFBP-1 gene methylation, gene polymorphism and serum levels of IGFBP-1. METHOD We included 100 subjects with T2DM and 100 control. DNA methylation of IGFBP-1 was analyzed using pyrosequencing, IGFBP-1 gene polymorphism was analyzed using real time polymerase chain reaction and serum level of IGFBP-1 was measured by ELISA. RESULTS There was DNA hyper methylation levels of IGFBP1 gene at each of the six CpG sites in T2DM patients than control (P < 0.001). IGFBP-1 gene polymorphism (rs 2854843) CC pattern was significantly associated with DM, P = 0.002. Also, there was decrease in serum IGFBP-1 in patients with T2DM than control group (P < 0.001). CONCLUSION We concluded that DNA hyper methylation of IGFBP-1 gene and CC polymorphism (rs 2854843) of IGFBP-1 gene are associated with T2DM in Egyptian patients, also, decrease serum level of IGFBP-1. Further cohort study is recommended with large sample size to detect which one, epigenetic changes or polymorphism of IGFBP-1 gene, is the cause of T2DM or even both.
Collapse
Affiliation(s)
- Nehal Salah Hasan
- Department of Clinical and Chemical Pathology, National Research Centre (NRC), Cairo, Egypt
| | - Hesham Gamal El Dine
- Department of Clinical and Chemical Pathology, National Research Centre (NRC), Cairo, Egypt
| | - Solaf Ahmed Kamel
- Department of Clinical and Chemical Pathology, National Research Centre (NRC), Cairo, Egypt
| | - Mona Hamed
- Department of Clinical and Chemical Pathology, National Research Centre (NRC), Cairo, Egypt
| | - Rasha N Youssef
- Department of Clinical and Chemical Pathology, National Research Centre (NRC), Cairo, Egypt
| | - Eman Mahmoud Hassan
- Department of Clinical and Chemical Pathology, National Research Centre (NRC), Cairo, Egypt.
| | | | - Asmaa Ali
- Department of pulmonary medicine, Abbassia Chest hospital, Ministry of health, Cairo, Egypt; Department of laboratory medicine, School of Medicine, Jiangsu University, Zhenjiang, P.R. China
| | - Eman Awadallah
- Department of Clinical and Chemical Pathology, National Research Centre (NRC), Cairo, Egypt
| |
Collapse
|
47
|
Binjawhar DN, Alhazmi AT, Bin Jawhar WN, MohammedSaeed W, Safi SZ. Hyperglycemia-induced oxidative stress and epigenetic regulation of ET-1 gene in endothelial cells. Front Genet 2023; 14:1167773. [PMID: 37139232 PMCID: PMC10150048 DOI: 10.3389/fgene.2023.1167773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction: Hyperglycemia-induced endothelial dysfunction and the subsequent increase of oxidative stress could lead to aberrant regulation of various genes which are responsible for a range of functions. This study aims to find out how hyperglycemia affect oxidative stress and then the expression and methylation of endothelin 1 (ET-1) gene in in human umbilical vein endothelial cells (HUVEC). Methods: Cells were cultured in growth medium and exposed to low and high glucose concentrations to mimic normal and diabetic condition respectively. Computational analysis were performed using UCSC genome browser and eukaryotic promoter database (EPD). The expression of ET-1 gene was investigated by real time PCR. Cytotoxicity and oxidative stress were determined by MTT and DCFH-DA assays respectively. Promoter methylation was assessed by the bisulfite sequencing method. Results: DCFH-DA assay showed that hyperglycemia can significantly increase the regulation of reactive oxygen species synthesis. The relative expression of ET-1 gene was increased due to exposure to high glucose concentration. MTT assay revealed reduced viability of cells due to the glucose induced damage. Methylation analysis revealed hypomethylation of the promoter of ET-1 however the difference was not significant. Out of 175 CpGs at 25 CpG sites, only 36 CpGs were methylated (20.5% methylation) in cell treated with normal glucose. Upon exposure to high glucose only 30 CpGs were methylated in 175 CpGs at 25 CpG sites (17.1% methylation). Discussion: Our study concludes a significantly high expression of ET-1 gene in response to high glucose exposure in HUVECs. It also reports that hyperglycemic condition leads to elevated oxidative stress. No significant change was found in methylation when cells were treated with high and low glucose concentrations.
Collapse
Affiliation(s)
- Dalal Nasser Binjawhar
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | | | - Wejdan Nasser Bin Jawhar
- Faculty—Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Walaa MohammedSaeed
- Department of Medical Laboratory Technology, Faculty of Applied Medical Science at Taibah University, Al Madinah Al Munawarah, Saudi Arabia
| | - Sher Zaman Safi
- Faculty of Medicine, Bioscience and Nursing, MAHSA University, Selangor, Malaysia
| |
Collapse
|
48
|
Kavian Z, Sargazi S, Majidpour M, Sarhadi M, Saravani R, Shahraki M, Mirinejad S, Heidari Nia M, Piri M. Association of SLC11A1 polymorphisms with anthropometric and biochemical parameters describing Type 2 Diabetes Mellitus. Sci Rep 2023; 13:6195. [PMID: 37062790 PMCID: PMC10106459 DOI: 10.1038/s41598-023-33239-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/10/2023] [Indexed: 04/18/2023] Open
Abstract
Diabetes, a leading cause of death globally, has different types, with Type 2 Diabetes Mellitus (T2DM) being the most prevalent one. It has been established that variations in the SLC11A1 gene impact risk of developing infectious, inflammatory, and endocrine disorders. This study is aimed to investigate the association between the SLC11A1 gene polymorphisms (rs3731864 G/A, rs3731865 C/G, and rs17235416 + TGTG/- TGTG) and anthropometric and biochemical parameters describing T2DM. Eight hundred participants (400 in each case and control group) were genotyped using the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) and amplification-refractory mutation system-PCR (ARMS-PCR) methods. Lipid profile, fasting blood sugar (FBS), hemoglobin A1c level, and anthropometric indices were also recorded for each subject. Findings revealed that SLC11A1-rs3731864 G/A, -rs17235416 (+ TGTG/- TGTG) were associated with T2DM susceptibility, providing protection against the disease. In contrast, SLC11A1-rs3731865 G/C conferred an increased risk of T2DM. We also noticed a significant association between SLC11A1-rs3731864 G/A and triglyceride levels in patients with T2DM. In silico evaluations demonstrated that the SLC11A2 and ATP7A proteins also interact directly with the SLC11A1 protein in Homo sapiens. In addition, allelic substitutions for both intronic variants disrupt or create binding sites for splicing factors and serve a functional effect. Overall, our findings highlighted the role of SLC11A1 gene variations might have positive (rs3731865 G/C) or negative (rs3731864 G/A and rs17235416 + TGTG/- TGTG) associations with a predisposition to T2DM.
Collapse
Affiliation(s)
- Zahra Kavian
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Mahdi Majidpour
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin Saravani
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mansour Shahraki
- Department of Nutrition, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
- Adolescent Health Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Milad Heidari Nia
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Maryam Piri
- Diabetes Center, Bu-Ali Hospital, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
49
|
Fernández-Carrión R, Sorlí JV, Asensio EM, Pascual EC, Portolés O, Alvarez-Sala A, Francès F, Ramírez-Sabio JB, Pérez-Fidalgo A, Villamil LV, Tinahones FJ, Estruch R, Ordovas JM, Coltell O, Corella D. DNA-Methylation Signatures of Tobacco Smoking in a High Cardiovascular Risk Population: Modulation by the Mediterranean Diet. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3635. [PMID: 36834337 PMCID: PMC9964856 DOI: 10.3390/ijerph20043635] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
Biomarkers based on DNA methylation are relevant in the field of environmental health for precision health. Although tobacco smoking is one of the factors with a strong and consistent impact on DNA methylation, there are very few studies analyzing its methylation signature in southern European populations and none examining its modulation by the Mediterranean diet at the epigenome-wide level. We examined blood methylation smoking signatures on the EPIC 850 K array in this population (n = 414 high cardiovascular risk subjects). Epigenome-wide methylation studies (EWASs) were performed analyzing differential methylation CpG sites by smoking status (never, former, and current smokers) and the modulation by adherence to a Mediterranean diet score was explored. Gene-set enrichment analysis was performed for biological and functional interpretation. The predictive value of the top differentially methylated CpGs was analyzed using receiver operative curves. We characterized the DNA methylation signature of smoking in this Mediterranean population by identifying 46 differentially methylated CpGs at the EWAS level in the whole population. The strongest association was observed at the cg21566642 (p = 2.2 × 10-32) in the 2q37.1 region. We also detected other CpGs that have been consistently reported in prior research and discovered some novel differentially methylated CpG sites in subgroup analyses. In addition, we found distinct methylation profiles based on the adherence to the Mediterranean diet. Particularly, we obtained a significant interaction between smoking and diet modulating the cg5575921 methylation in the AHRR gene. In conclusion, we have characterized biomarkers of the methylation signature of tobacco smoking in this population, and suggest that the Mediterranean diet can increase methylation of certain hypomethylated sites.
Collapse
Affiliation(s)
- Rebeca Fernández-Carrión
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José V. Sorlí
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva M. Asensio
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eva C. Pascual
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Olga Portolés
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Andrea Alvarez-Sala
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Francesc Francès
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Alejandro Pérez-Fidalgo
- Department of Medical Oncology, University Clinic Hospital of Valencia, 46010 Valencia, Spain
- Biomedical Research Networking Centre on Cancer (CIBERONC), Health Institute Carlos III, 28029 Madrid, Spain
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
| | - Laura V. Villamil
- Department of Physiology, School of Medicine, University Antonio Nariño, Bogotá 111511, Colombia
| | - Francisco J. Tinahones
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, 29590 Málaga, Spain
| | - Ramon Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Jose M. Ordovas
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA
- Nutritional Control of the Epigenome Group, Precision Nutrition and Obesity Program, IMDEA Food, UAM + CSIC, 28049 Madrid, Spain
| | - Oscar Coltell
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Computer Languages and Systems, Universitat Jaume I, 12071 Castellón, Spain
| | - Dolores Corella
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
50
|
Sibuh BZ, Quazi S, Panday H, Parashar R, Jha NK, Mathur R, Jha SK, Taneja P, Jha AK. The Emerging Role of Epigenetics in Metabolism and Endocrinology. BIOLOGY 2023; 12:256. [PMID: 36829533 PMCID: PMC9953656 DOI: 10.3390/biology12020256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023]
Abstract
Each cell in a multicellular organism has its own phenotype despite sharing the same genome. Epigenetics is a somatic, heritable pattern of gene expression or cellular phenotype mediated by structural changes in chromatin that occur without altering the DNA sequence. Epigenetic modification is an important factor in determining the level and timing of gene expression in response to endogenous and exogenous stimuli. There is also growing evidence concerning the interaction between epigenetics and metabolism. Accordingly, several enzymes that consume vital metabolites as substrates or cofactors are used during the catalysis of epigenetic modification. Therefore, altered metabolism might lead to diseases and pathogenesis, including endocrine disorders and cancer. In addition, it has been demonstrated that epigenetic modification influences the endocrine system and immune response-related pathways. In this regard, epigenetic modification may impact the levels of hormones that are important in regulating growth, development, reproduction, energy balance, and metabolism. Altering the function of the endocrine system has negative health consequences. Furthermore, endocrine disruptors (EDC) have a significant impact on the endocrine system, causing the abnormal functioning of hormones and their receptors, resulting in various diseases and disorders. Overall, this review focuses on the impact of epigenetics on the endocrine system and its interaction with metabolism.
Collapse
Affiliation(s)
- Belay Zeleke Sibuh
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Sameer Quazi
- GenLab Biosolutions Private Limited, Bangalore 560043, India
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge CB1 1PT, UK
- Clinical Bioinformatics, School of Health Sciences, The University of Manchester, Manchester M13 9P, UK
- SCAMT Institute, ITMO University, St. Petersburg 197101, Russia
| | - Hrithika Panday
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Ritika Parashar
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India
| | - Runjhun Mathur
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali 140413, India
| | - Pankaj Taneja
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| | - Abhimanyu Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Knowledge Park III, Greater Noida 201310, India
| |
Collapse
|